1
|
Bento R, Scheller J, Parekkadan B. Intratumoral Delivery of Genetically Engineered Anti-IL-6 Trans-signaling Therapeutics. Mol Biotechnol 2025; 67:2696-2708. [PMID: 38980514 PMCID: PMC12119671 DOI: 10.1007/s12033-024-01230-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 06/17/2024] [Indexed: 07/10/2024]
Abstract
Interleukin-6 (IL-6) is a highly pro-inflammatory cytokine involved in the etiopathology of several inflammatory diseases and cancer. As so, the inhibition of IL-6 signaling pathways has emerged as an attractive therapeutic avenue for the treatment of several chronic diseases. Since IL-6 trans-signaling was described as the pathological branch of IL-6, selective inhibitors were developed. Next-generation variants with increased trans-signaling specificity and potency emerged as great candidates for the treatment of several diseases, with reduced off-target effects. The highly time-consuming and costly processes involving recombinant protein production, however, have hampered the progress of anti-cytokine pharmaceuticals in clinic so far. Herein, we developed gene therapeutic modalities of IL-6-trans-signaling inhibitors as alternatives for sustained recombinant protein secretion. By using an IL-6-dependent lymphoma cell line and xenograft tumor model, we demonstrated the superior inhibitory potential of second-generation anti-IL-6 trans-signaling therapeutic. We compared the efficiency of distinct gene delivery modalities using a bioluminescent biomarker probe and observed consistent protein production via cell-based delivery. When delivered intratumorally, genetically engineered sgp130FlyRFc-secreting cells significantly reduced tumor burden and increased animal survival, representing a promising therapeutic avenue to be explored in clinically relevant gene delivery applications.
Collapse
Affiliation(s)
- Raphaela Bento
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA
| | - Jürgen Scheller
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Biju Parekkadan
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA.
| |
Collapse
|
2
|
Amer H, Flanagan KL, Kampan NC, Itsiopoulos C, Scott CL, Kartikasari AER, Plebanski M. Interleukin-6 Is a Crucial Factor in Shaping the Inflammatory Tumor Microenvironment in Ovarian Cancer and Determining Its Hot or Cold Nature with Diagnostic and Prognostic Utilities. Cancers (Basel) 2025; 17:1691. [PMID: 40427188 PMCID: PMC12109964 DOI: 10.3390/cancers17101691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 05/05/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025] Open
Abstract
Ovarian cancer (OC) remains the leading cause of cancer-related deaths among women, often diagnosed at advanced stages due to the lack of effective early diagnostic procedures. To reduce the high mortality rates in OC, reliable biomarkers are urgently needed, especially to detect OC at its earliest stage, predict specific drug responses, and monitor patients. The cytokine interleukin-6 (IL6) is associated with low survival rates, treatment resistance, and recurrence. In this review, we summarize the role of IL6 in inflammation and how IL6 contributes to ovarian tumorigenesis within the tumor microenvironment, influencing whether the tumor is subsequently classified as "hot" or "cold". We further dissect the molecular and cellular mechanisms through which IL6 production and downstream signaling are regulated, to enhance our understanding of its involvement in OC development, as well as OC resistance to treatment. We highlight the potential of IL6 to be used as a reliable diagnostic biomarker to help detect OC at its earliest stage, and as a part of predictive and prognostic signatures to improve OC management. We further discuss ways to leverage artificial intelligence and machine learning to integrate IL6 into diverse biomarker-based strategies.
Collapse
Affiliation(s)
- Hina Amer
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3082, Australia
| | - Katie L. Flanagan
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3082, Australia
- School of Medicine and Health Sciences, University of Tasmania, Launceston, TAS 7250, Australia
- Tasmanian Vaccine Trial Centre, Clifford Craig Foundation, Launceston General Hospital, Launceston, TAS 7250, Australia
| | - Nirmala C. Kampan
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Catherine Itsiopoulos
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3082, Australia
| | - Clare L. Scott
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Faculty of Medicine, Dentistry, and Health Sciences, The University of Melbourne, Parkville, VIC 3052, Australia
- The Royal Women’s Hospital, Parkville, VIC 3052, Australia
| | | | - Magdalena Plebanski
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3082, Australia
| |
Collapse
|
3
|
Iwanczyk Z, Hara H, Cooper DKC, Maenaka A. Inhibition of inflammation by IL-6 blockade in xenotransplantation. Cytokine 2025; 189:156897. [PMID: 39999679 PMCID: PMC11976666 DOI: 10.1016/j.cyto.2025.156897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/23/2025] [Accepted: 02/16/2025] [Indexed: 02/27/2025]
Abstract
The inflammatory cytokine interleukin 6 (IL-6) plays a role in both acute and chronic organ allotransplant rejection. Data suggest that IL-6 inhibition may help prevent or reverse rejection, with large multi-center trials now underway. However, the evidence for the benefit of IL-6 inhibitors in xenotransplantation is limited. IL-6 inhibition has been explored in nonhuman-primate models of xenotransplantation, but no clear consensus exists on its efficacy or the best mode of IL-6 inhibition (anti-IL-6 antibodies, or through IL-6 receptor [IL-6R] blockade). Extra considerations for IL-6 blockade exist in xenotransplantation, as both recipient (human) and xenograft-derived (porcine) IL-6 may play roles. The systemic inflammation seen in xenograft recipients (SIXR) contributes to significant morbidity and mortality for the recipient through coagulation dysfunction and augmentation of the immune response. Anti-IL-6 antibodies (e.g., siltuximab) bind to human IL-6 and prevent IL-6R activation, but do not bind to porcine IL-6, and so have no effect in preventing graft-driven inflammatory processes. In contrast, IL-6R inhibitors (e.g., tocilizumab) inhibit IL-6 activity by blocking binding of human and porcine IL-6 to human IL-6R. Although IL-6R blockade cannot prevent the effect of IL-6 on porcine cells, it probably prevents graft-derived IL-6 from contributing to an inflammatory response in the host. This review outlines the role of IL-6 in xenotransplantation and discusses mechanisms for inhibiting IL-6 to improve recipient survival.
Collapse
Affiliation(s)
- Zuzanna Iwanczyk
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Hidetaka Hara
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, Yunnan, China
| | - David K C Cooper
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Akihiro Maenaka
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
4
|
Ducharme JB, Specht JW, Bailly AR, Deyhle MR. Serum cytokines and their soluble receptors are differently regulated between trained and untrained men after vigorous endurance exercise. Am J Physiol Regul Integr Comp Physiol 2025; 328:R581-R587. [PMID: 40130752 DOI: 10.1152/ajpregu.00010.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/02/2025] [Accepted: 03/14/2025] [Indexed: 03/26/2025]
Abstract
Endurance training contributes to immune system changes that help manage exercise-induced stress and promote an anti-inflammatory effect. However, the specific mechanisms underlying these adaptations are still being explored. Cytokines play a key role in both acute and chronic exercise responses through interactions with their receptors, which are present in both membrane-bound and soluble forms. Yet, the impact of exercise on cytokines and soluble cytokine receptors in parallel remains understudied. Therefore, the purpose of this study was to assess how cytokines and their soluble receptors change in serum after vigorous exercise in endurance-trained and untrained men. Following 1-h of cycling at their respiratory compensation point, untrained men (n = 5) exhibited a significant increase in proinflammatory cytokines, including IL-6, TNF-α, IL-1β, CCL2, and VEGFA. In contrast, anti-inflammatory cytokines such as IL-10 and IL-1Ra were reduced. These effects were not observed in the trained group (n = 7). Instead, proinflammatory cytokine levels remained close to the baseline, whereas the anti-inflammatory cytokines IL-10 and IL-1Ra increased. In the trained group, these cytokine changes were accompanied by a marked increase in the expression of soluble cytokine receptors known to inhibit cytokine-mediated signaling, such as sIL-1RII, sGP130, sTNFRI, sTNFRII, sVEGFR1, and sVEGFR2, indicating reduced cytokine bioavailability. However, in the untrained group, the expression of these soluble cytokine receptors either remained unchanged or decreased, suggesting greater cytokine bioavailability. Together these findings highlight a novel potential anti-inflammatory adaptation such that trained men present a blunted inflammatory response by both reduced inflammatory cytokines and increased soluble cytokine receptors after exercise compared with untrained men.NEW & NOTEWORTHY Our study identifies a novel potential anti-inflammatory adaptation to endurance training, where trained men present a blunted inflammatory response characterized by reduced inflammatory cytokines and increased soluble cytokine receptors, resulting in decreased cytokine bioavailability after exercise compared with untrained men.
Collapse
Affiliation(s)
- Jeremy B Ducharme
- Department of Health, Exercise and Sports Sciences, University of New Mexico, Albuquerque, New Mexico, United States
| | - Jonathan W Specht
- Department of Health, Exercise and Sports Sciences, University of New Mexico, Albuquerque, New Mexico, United States
| | - Alyssa R Bailly
- Department of Health, Exercise and Sports Sciences, University of New Mexico, Albuquerque, New Mexico, United States
| | - Michael R Deyhle
- Department of Health, Exercise and Sports Sciences, University of New Mexico, Albuquerque, New Mexico, United States
- Department of Cell Biology and Physiology, School of Medicine, University of New Mexico, Albuquerque, New Mexico, United States
| |
Collapse
|
5
|
Wunderlich F, Gerovska D, Delic D, Araúzo-Bravo MJ. Protective Vaccination of Mice Against Blood-Stage Malaria Impacts Hepatic Expression of Genes Encoding Acute-Phase Proteins and IL-6 Family Members. Int J Mol Sci 2025; 26:3173. [PMID: 40243929 PMCID: PMC11989154 DOI: 10.3390/ijms26073173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/25/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
In response to vaccination and/or infectious agents, the liver produces acute-phase proteins (APPs) driven by IL-6, which circulate in blood plasma as components of the humoral innate defense. This study investigates the liver of mice for possible effects of protective vaccination against primary blood-stage infections of Plasmodium chabaudi malaria on the expression of genes encoding APPs and IL-6 family members. Female Balb/c mice were vaccinated with a non-infectious vaccine prior to challenge with 106P. chabaudi-infected erythrocytes, resulting in about 80% survival of otherwise lethal infections. Gene expression microarrays were used to determine the relative transcript levels of genes in the livers of vaccinated and unvaccinated mice on days 0, 1, 4, 8, and 11 p.i. (post infectionem). Vaccination induced significant (p-value < 0.05) differences in the expression of malaria-responsive genes toward the end of crisis on day 11 p.i., when mice recovered from infections. These genes include Saa4, Apcs, Cp, and Crp, encoding APPs described to inhibitorily interact with parasitic blood stages; the genes F2, F7, F8, F9, F10, and F13b, and Plg, Plat, and Serpina5, encoding proteins balancing coagulation vs. fibrinolysis dysregulated by malaria, respectively; the genes Hc, C8a, C8b, C8g, and C9, encoding components of lytic complement membrane attack complex (MAC); and Cfh, Cfi, and C4bp, encoding complement-regulatory proteins. Vaccination accelerated, albeit differently, the malaria-induced activation of all three complement pathways, evidenced as higher transcript levels of C1qa, C1qb, C1qc, Fcna, Cfp, C3, Cfh, C8a, and C9 on day 4 p.i., C1ra, C1s, and C2 on day 1 p.i., and Serping1, encoding the multifunctional protease inhibitor C1INH, on day 0 p.i. Protective vaccination may also accelerate downregulation of the malaria-promoting lethality of IL-6 trans-signaling, which may contribute to an overall accelerated recovery of mice from otherwise lethal blood-stage malaria.
Collapse
Affiliation(s)
- Frank Wunderlich
- Department of Biology, Heinrich-Heine-University, 40225 Düsseldorf, Germany;
| | - Daniela Gerovska
- Computational Biology and Systems Biomedicine, Biogipuzkoa Health Research Institute, 20014 San Sebastian, Spain;
| | - Denis Delic
- Boehringer Ingelheim Pharma & Co., KG, 88400 Biberach, Germany
| | - Marcos J. Araúzo-Bravo
- Computational Biology and Systems Biomedicine, Biogipuzkoa Health Research Institute, 20014 San Sebastian, Spain;
- IKERBASQUE, Basque Foundation for Science, 48009 Bilbao, Spain
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of Basque Country (UPV/EHU), 48940 Leioa, Spain
| |
Collapse
|
6
|
Lospinoso Severini F, Falco G, Notarangelo T. Role of Soluble Cytokine Receptors in Gastric Cancer Development and Chemoresistance. Int J Mol Sci 2025; 26:2534. [PMID: 40141175 PMCID: PMC11942508 DOI: 10.3390/ijms26062534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 02/28/2025] [Accepted: 03/08/2025] [Indexed: 03/28/2025] Open
Abstract
Gastric cancer is among the top five most important malignancies in the world due to the high burden of the disease and its lethality. Indeed, it is the fourth most common cause of death worldwide, characterized by a poor prognosis and low responsiveness to chemotherapy. Multidrug resistance limits the clinical management of the patient. Among these, the role of chronic activation of inflammatory pathways underlying gastric tumorigenesis should be highlighted. Furthermore, the gastric immunosuppressive TME influences the response to therapy. This review discusses the role of soluble cytokine receptors in the development and chemoresistance of gastric cancer, considered as a molecular marker and target of strategies to overcome resistance.
Collapse
Affiliation(s)
- Francesca Lospinoso Severini
- Laboratory of Preclinical and Translational Research, IRCCS CROB Centro di Riferimento Oncologico della Basilicata, 85028 Rionero in Vulture, PZ, Italy
| | - Geppino Falco
- Department of Biology, University of Naples Federico II, 80138 Napoli, NA, Italy
- Biogem, Istituto di Biologia e Genetica Molecolare, 83031 Ariano Irpino, AV, Italy
| | - Tiziana Notarangelo
- Laboratory of Preclinical and Translational Research, IRCCS CROB Centro di Riferimento Oncologico della Basilicata, 85028 Rionero in Vulture, PZ, Italy
| |
Collapse
|
7
|
Hall C, Nguyen DT, Mendoza K, Tan C, Chauhan A. Inhibition of IL-6 trans-signaling promotes post-stroke functional recovery in a sex and dose-dependent manner. J Neuroinflammation 2025; 22:52. [PMID: 40011978 PMCID: PMC11866694 DOI: 10.1186/s12974-025-03365-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 02/01/2025] [Indexed: 02/28/2025] Open
Abstract
INTRODUCTION Elevated circulating IL-6 levels are associated with poorer outcomes after stroke, and increased serum IL-6 levels are linked to a higher risk of stroke. IL-6 binds to soluble IL-6 receptors (sIL-6R) and subsequently to ubiquitously expressed gp130, initiating proinflammatory trans-signaling. This study tested the hypothesis that inhibiting IL-6 trans-signaling by administering soluble (s) gp130 improves long-term functional outcomes in young mice after stroke. METHODS Recombinant mouse gp130Fc chimera (sgp130) was administered one hour after middle cerebral artery occlusion (MCAO) followed by twice-weekly administration for 2 weeks in mice (8-15 weeks old). Behavioral assessments were done on days 7 and 28 post-MCAO for chronic studies. Flow cytometry was performed on days 3 (blood) and 7 (spleen and brain) to assess IL-6, mIL-6R, and phosphorylated STAT3 expression. RESULTS Improved long-term functional outcomes were observed in male, but not female mice. To investigate the differential response in females, ELISA analyses revealed that plasma IL-6 levels increased in both sexes after MCAO, with a more pronounced induction in females. Additionally, circulating sIL-6R levels were significantly higher in females compared to males (p < 0.05) at 24 h post-MCAO. Administering a higher dose of sgp130 (1 mg/kg) to females improved long-term functional outcomes, suggesting that a higher dose is needed to inhibit IL-6 trans-signaling in females effectively. Mechanistically, sgp130 treatment reduced phosphorylated STAT3 expression in brain F4/80 macrophages and increased the expression of mIL-6R on splenic immune cells at day 7 post-MCAO in both sexes. CONCLUSION These findings demonstrate that inhibition of IL-6 trans-signaling with gp130Fc improves long-term functional outcomes in both male and female mice, albeit in a dose-dependent manner. This study provides novel insights into potential therapeutic strategies targeting IL-6 signaling pathways following stroke.
Collapse
Affiliation(s)
- Cassandra Hall
- Department of Neurology, University of Texas McGovern Medical School at Houston, Houston, TX, USA
| | - Dustin T Nguyen
- Department of Neurology, University of Texas McGovern Medical School at Houston, Houston, TX, USA
| | - Kate Mendoza
- Department of Neurology, University of Texas McGovern Medical School at Houston, Houston, TX, USA
| | - Chunfeng Tan
- Department of Neurology, University of Texas McGovern Medical School at Houston, Houston, TX, USA
| | - Anjali Chauhan
- Department of Neurology, University of Texas McGovern Medical School at Houston, Houston, TX, USA.
| |
Collapse
|
8
|
Cao J, Du T, He K, Dai Z, Xie X, Chen B, Feng J, Xu T. Mendelian randomization and colocalization analysis reveal the role of IL-6 signaling pathway in psoriasis and potential therapeutic targets. Arch Dermatol Res 2025; 317:468. [PMID: 39987260 DOI: 10.1007/s00403-025-03995-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/30/2025] [Accepted: 02/03/2025] [Indexed: 02/24/2025]
Abstract
IL-6 signaling is critical in the pathogenesis of several autoimmune diseases, including psoriasis and psoriatic arthritis. This study utilized Mendelian randomization (MR) analysis to investigate the genetic associations between IL-6 signaling, soluble IL-6 receptor (sIL-6R) levels, and psoriasis and its arthritis. Additionally, the therapeutic potential of IL-6R inhibitors was evaluated. The study methodology was validated through positive and negative control analysis, with further confirmation of genetic associations via Bayesian factor colocalization analysis. The MR results indicated that upregulation of IL-6 signaling was positively associated with psoriasis (PIVW = 0.008, 95% CI: 1.16-2.66), whereas elevated sIL-6R levels were negatively associated with psoriasis (PIVW < 0.001, 95% CI: 0.95-0.99). Positive control analysis showed that IL-6R inhibitors effectively reduced the risk of rheumatoid arthritis but increased the risk of atopic dermatitis. In contrast, negative control analysis revealed no significant therapeutic effect on seborrheic dermatitis. Using GWAS data from multiple databases, IL-6R inhibitors were identified as being associated with a reduced risk of psoriasis and psoriatic arthritis. Colocalization analysis highlighted the rs4129267 genetic variant as strongly linked to the therapeutic effects of IL-6R inhibitors on psoriasis (P < 0.001, 95% CI: 0.37-0.74). These findings underscore the crucial role of IL-6 signaling in the development of psoriasis and psoriatic arthritis and provide robust genetic evidence supporting the therapeutic potential of IL-6R inhibitors. By identifying genetic determinants of disease susceptibility and potential biomarkers of treatment response, this study provides valuable clinical insights and guidance for optimizing treatment strategies for psoriasis.
Collapse
Affiliation(s)
- Jianye Cao
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Tiantao Du
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Kaiming He
- Department of Thoracic Surgery, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Ziyao Dai
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Xianting Xie
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Baiyu Chen
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Jia Feng
- Department of Laboratory Medicine, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| | - Tao Xu
- Department of Thoracic Surgery, Affiliated Hospital of Southwest Medical University, Luzhou, China.
| |
Collapse
|
9
|
Abdelhamed HG, Hassan AA, Sakraan AA, Al-Deeb RT, Mousa DM, Aboul Ezz HS, Noor NA, Khadrawy YA, Radwan NM. Brain interleukins and Alzheimer's disease. Metab Brain Dis 2025; 40:116. [PMID: 39891777 PMCID: PMC11787210 DOI: 10.1007/s11011-025-01538-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 01/10/2025] [Indexed: 02/03/2025]
Abstract
The central nervous system (CNS) is immune-privileged by several immuno-modulators as interleukins (ILs). ILs are cytokines secreted by immune cells for cell-cell signaling communications and affect the functions of the CNS. ILs were reported to orchestrate different molecular and cellular mechanisms of both physiological and pathological events, through overproduction or over-expression of their receptors. They interact with numerous receptors mediating pro-inflammatory and/or anti-inflammatory actions. Interleukins have been implicated to participate in neurodegenerative diseases. They play a critical role in Alzheimer's disease (AD) pathology which is characterized by the over-production of pro-inflammatory ILs. These may aggravate neurodegeneration, in addition to their contribution to detrimental mechanisms as oxidative stress, and excitotoxicity. However, recent research on the relation between ILs and AD revealed major discrepancies. Most of the major ILs were shown to play both pro- and anti-inflammatory roles in different experimental settings and models. The interactions between different ILs through shared pathways also add to the difficulty of drawing solid conclusions. In addition, targeting the different ILs has not yielded consistent results. The repeated failures of therapeutic drugs in treating AD necessitate the search for novel agents targeting multiple mechanisms of the disease pathology. In this context, the understanding of interleukins and their roles throughout the disease progression and interaction with other systems in the brain may provide promising therapeutic targets for the prevention or treatment of AD.
Collapse
Affiliation(s)
- Heba G Abdelhamed
- Department of Zoology and Chemistry, Faculty of Science, Cairo University, Giza, Egypt
| | - Arwa A Hassan
- Faculty of Pharmacy & Pharmaceutical Industries, Sinai University, Sinai, Egypt
| | - Alaa A Sakraan
- Department of Zoology, Faculty of Science, Cairo University, Giza, Egypt
| | | | - Dalia M Mousa
- Department of Biotechnology, Faculty of Science, Cairo University, Giza, Egypt
| | - Heba S Aboul Ezz
- Department of Zoology, Faculty of Science, Cairo University, Giza, Egypt.
| | - Neveen A Noor
- Department of Zoology, Faculty of Science, Cairo University, Giza, Egypt
| | - Yasser A Khadrawy
- Medical Physiology Department, Medical Research and Clinical Studies Institute, National Research Center, Giza, Egypt
| | - Nasr M Radwan
- Department of Zoology, Faculty of Science, Cairo University, Giza, Egypt
| |
Collapse
|
10
|
Weitz HT, Ettich J, Rafii P, Wittich C, Schultz L, Frank NC, Heise D, Krusche M, Lokau J, Garbers C, Behnke K, Floss DM, Kolmar H, Moll JM, Scheller J. Interleukin-11 receptor is an alternative α-receptor for interleukin-6 and the chimeric cytokine IC7. FEBS J 2025; 292:523-536. [PMID: 39473075 PMCID: PMC11796321 DOI: 10.1111/febs.17309] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 07/30/2024] [Accepted: 10/16/2024] [Indexed: 02/06/2025]
Abstract
The cytokine interleukin 6 (IL-6) signals via the IL-6 α-receptor (IL-6Rα or IL-6R) in complex with the gp130 β-receptor. Cell type restricted expression of the IL-6R limits the action of IL-6 mainly to hepatocytes and some immune cells. Here, we show that IL-6 also binds to the IL-11 α receptor (IL-11Rα or IL-11R) and induces signaling via IL-11R:gp130 complexes, albeit with a lower affinity compared to IL-11. Antagonistic antibodies directed against IL-11R, but not IL-6R, inhibit IL-6 signaling via IL-11R:gp130 receptor complexes. Notably, IL-11 did not cross-react with IL-6R. IL-11R has also been identified as an alternative α receptor for the CNTF/IL-6-derived chimeric cytokine IC7, which has recently been shown to induce weight loss in mice. Accordingly, the effects of therapeutic monoclonal antibodies against IL-6 or IL-6R, which both block IL-6 signaling, may be slightly different. These findings provide new insights into IL-6 signaling and therefore offer new potential therapeutic intervention options in the future.
Collapse
Affiliation(s)
- Hendrik T. Weitz
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital DüsseldorfHeinrich Heine University DüsseldorfGermany
| | - Julia Ettich
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital DüsseldorfHeinrich Heine University DüsseldorfGermany
| | - Puyan Rafii
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital DüsseldorfHeinrich Heine University DüsseldorfGermany
| | - Christoph Wittich
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital DüsseldorfHeinrich Heine University DüsseldorfGermany
| | - Laura Schultz
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital DüsseldorfHeinrich Heine University DüsseldorfGermany
| | - Nils C. Frank
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital DüsseldorfHeinrich Heine University DüsseldorfGermany
| | - Denise Heise
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital DüsseldorfHeinrich Heine University DüsseldorfGermany
| | - Matthias Krusche
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital DüsseldorfHeinrich Heine University DüsseldorfGermany
| | - Juliane Lokau
- Institute of Clinical BiochemistryHannover Medical SchoolGermany
| | | | - Kristina Behnke
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital DüsseldorfHeinrich Heine University DüsseldorfGermany
| | - Doreen M. Floss
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital DüsseldorfHeinrich Heine University DüsseldorfGermany
| | - Harald Kolmar
- Institute for Organic Chemistry and BiochemistryTechnical University of DarmstadtGermany
- Centre of Synthetic BiologyTechnical University of DarmstadtGermany
| | - Jens M. Moll
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital DüsseldorfHeinrich Heine University DüsseldorfGermany
| | - Jürgen Scheller
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital DüsseldorfHeinrich Heine University DüsseldorfGermany
| |
Collapse
|
11
|
Komaki S, Inagaki T, Kumar A, Izumiya Y. The Role of vIL-6 in KSHV-Mediated Immune Evasion and Tumorigenesis. Viruses 2024; 16:1900. [PMID: 39772207 PMCID: PMC11680145 DOI: 10.3390/v16121900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 12/05/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) is a double-stranded DNA gamma herpesvirus. Like other herpesviruses, KSHV establishes a latent infection with limited gene expression, while KSHV occasionally undergoes the lytic replication phase, which produces KSHV progenies and infects neighboring cells. KSHV genome encodes 80+ open reading frames. One of the KSHV genes, K2, encodes viral interleukin 6 (vIL-6), a homolog of human IL-6 (hIL-6), mainly expressed in the lytic phase of the virus. vIL-6 plays a crucial role in regulating the expression of other viral genes and is also associated with inducing angiogenesis, cell survival, and immune evasion, which is suggested to promote the development of KSHV-associated diseases. This review summarizes the current knowledge on vIL-6. We focus on the vIL-6 regarding its protein structure, transcriptional regulation, cell signaling pathways, and contribution to the KSHV-associated diseases.
Collapse
Affiliation(s)
- Somayeh Komaki
- Department of Dermatology, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Tomoki Inagaki
- Department of Dermatology, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Ashish Kumar
- Department of Dermatology, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Yoshihiro Izumiya
- Department of Dermatology, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
| |
Collapse
|
12
|
Seibel C, Pudewell S, Rafii P, Ettich J, Weitz HT, Lang A, Petzsch P, Köhrer K, Floss DM, Scheller J. Synthetic trimeric interleukin-6 receptor complexes with a STAT3 phosphorylation dominated activation profile. Cytokine 2024; 184:156766. [PMID: 39348731 DOI: 10.1016/j.cyto.2024.156766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/11/2024] [Accepted: 09/18/2024] [Indexed: 10/02/2024]
Abstract
In Interleukin (IL)-6 signalling, IL-6 site I binds to the IL-6 receptor (IL-6R) first, following by IL-6 site II interaction to domain 2/3 of gp130 to form premature trimeric IL-6:IL-6R:gp130 receptor complexes. Formation of the mature hexameric receptor complex is then facilitated by the inter-trimeric interaction of IL-6 site III with domain 1 of the opposing gp130. The two gp130-associated Janus kinases (JAKs) trans-phosphorylate when their spatiotemporal pairing is correct, which causes the activation of STAT, ERK, and AKT pathways in a balanced manner. Since the intracellular domain (ICD) of IL-6R is not needed for STAT/ERK/AKT phosphorylation, we investigated the conditions under which a chimeric IL-6RECD-gp130TMD/ICD receptor protein confers biological activity. For IL-6RECD-gp130TMD/ICD, the extracellular domain (ECD) of IL-6R was fused to the transmembrane domain (TMD) and ICD of gp130. Co-expression of IL-6RECD-gp130TMD/ICD with signalling-deficient gp130 variants did not induce IL-6 signalling, suggesting that the assembly of hexameric complexes failed to dimerize the IL-6R-associated JAKs correctly. By mimicking the premature trimeric receptor complex, IL-6-mediated dimerization of IL-6RECD-gp130TMD/ICD with the single-cytokine-binding variant gp130ΔD1 induced signalling. Of note, IL-6 signalling via these synthetic gp130ΔD1:IL-6RECD-gp130TMD/ICD complexes resulted predominantly in STAT3 phosphorylation. A STAT3-dominated profile was also observed after IL-6-induced signalling mediated by a JAK-deficient IL-6RECD-gp130TMD/ICDΔJAK variant in complex with the JAK-proficient but STAT/ERK/AKT-deficient gp130JAKΔICD variant. Our data showed that effective ERK/AKT signalling could not be executed after intracellular domain swapping from gp130 to the IL-6R. Taken together, the chimeric IL-6R/gp130 receptor may be helpful in the creation of customized synthetic IL-6 signalling.
Collapse
Affiliation(s)
- Christiane Seibel
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
| | - Silke Pudewell
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
| | - Puyan Rafii
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
| | - Julia Ettich
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
| | - Hendrik T Weitz
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
| | - Alexander Lang
- Cardiovascular Research Laboratory, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
| | - Patrick Petzsch
- Biological and Medical Research Center (BMFZ), Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
| | - Karl Köhrer
- Biological and Medical Research Center (BMFZ), Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
| | - Doreen M Floss
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
| | - Jürgen Scheller
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany.
| |
Collapse
|
13
|
Curren B, Ahmed T, Rashid RB, Sebina I, Al Amin Sikder M, Howard DR, Alorro M, Ullah MA, Bissell A, Rahman MM, Pearen MA, Ramm GA, Varelias A, Rose-John S, MacDonald KPA, Hoelzle R, Ó Cuív P, Spann KM, Dennis PG, Phipps S. A maternal high-fat diet predisposes to infant lung disease via increased neutrophil-mediated IL-6 trans-signaling. Cell Rep 2024; 43:114974. [PMID: 39535919 DOI: 10.1016/j.celrep.2024.114974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 09/24/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
A poor maternal diet during pregnancy predisposes the infant to severe lower respiratory tract infections (sLRIs), which, in turn, increases childhood asthma risk; however, the underlying mechanisms remain poorly understood. Here, we show that the offspring of high-fat diet (HFD)-fed mothers (HFD-reared pups) developed an sLRI following pneumovirus inoculation in early life and subsequent asthma in later life upon allergen exposure. Prior to infection, HFD-reared pups developed microbial dysbiosis and low-grade systemic inflammation (LGSI), characterized by hyperneutropoiesis in the liver and elevated inflammatory cytokine expression, most notably granulocyte-colony stimulating factor (G-CSF), interleukin-17A (IL-17A), IL-6 and soluble IL-6 receptor (sIL-6R) (indicative of IL-6 trans-signaling) in the circulation and multiple organs but most prominently the liver. Inhibition of IL-6 trans-signaling using sgp130Fc transgenic mice or via specific genetic deletion of IL-6Ra on neutrophils conferred protection against both diseases. Taken together, our findings suggest that a maternal HFD induces neonatal LGSI that predisposes to sLRI and subsequent asthma via neutrophil-mediated IL-6 trans-signaling.
Collapse
Affiliation(s)
- Bodie Curren
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, QLD 4072, Australia
| | - Tufael Ahmed
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, QLD 4000, Australia
| | - Ridwan B Rashid
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
| | - Ismail Sebina
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
| | - Md Al Amin Sikder
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, QLD 4072, Australia
| | - Daniel R Howard
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, QLD 4072, Australia
| | - Mariah Alorro
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
| | - Md Ashik Ullah
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
| | - Alec Bissell
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
| | - Muhammed Mahfuzur Rahman
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, QLD 4072, Australia
| | - Michael A Pearen
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
| | - Grant A Ramm
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, QLD 4072, Australia
| | - Antiopi Varelias
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, QLD 4000, Australia; Australian Infectious Diseases Research Centre, The University of Queensland, QLD 4072, Australia
| | - Stefan Rose-John
- Christian-Albrechts-Universität zu Kiel, Medical Faculty, Olshausenstraße 40, 24098 Kiel, Germany
| | - Kelli P A MacDonald
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, QLD 4072, Australia
| | - Robert Hoelzle
- School of Environment, The University of Queensland, QLD 4072, Australia
| | - Páraic Ó Cuív
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, QLD 4000, Australia
| | - Kirsten M Spann
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, QLD 4000, Australia
| | - Paul G Dennis
- Australian Infectious Diseases Research Centre, The University of Queensland, QLD 4072, Australia; School of Environment, The University of Queensland, QLD 4072, Australia
| | - Simon Phipps
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, QLD 4072, Australia; School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, QLD 4000, Australia; Australian Infectious Diseases Research Centre, The University of Queensland, QLD 4072, Australia.
| |
Collapse
|
14
|
Zhang Z, Shi D, Dou H, Wan R, Yuan Q, Tu P, Xin D. Mycoplasma pneumoniae regulates the expression of GP130 in lung epithelial cells through apoptosis and TLR4/ NF-κB pathway during infection. Microb Pathog 2024; 197:107072. [PMID: 39447660 DOI: 10.1016/j.micpath.2024.107072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 10/12/2024] [Accepted: 10/21/2024] [Indexed: 10/26/2024]
Abstract
In previous study, lower levels of serum GP130 were reported in children with MPP. GP130 is an important signal transducer, the down regulation of which may influence host immune responses. In this study, we aimed to analyze the regulatory mechanism of GP130 during MP infection. Firstly, the mRNA and protein levels of GP130 both decrease and then increase with increasing multiplicity of infection (MOI: 1 to 40) of MP. The lowest levels of GP130 were detected at MOI of 5. Then, heat treated MP but not trypsin treated MP or MP extracted proteins show regulatory effect to the expression of GP130. These indicate that the down regulation of GP130 is related to protein mediate adhesion process of MP. Gene expression analysis revealed that MP affected apoptosis and the TLR4 pathway in infected cells, and the mRNA level of IL-6 was correlated with that of GP130. Further, Z-VAD-FMK (pan-caspase inhibitor) can suppress the apoptosis induced by MP infection and restore GP130 at protein level. Further studies revealed that MP infection promoted TLR4 internalization but did not activate the NF-κB pathway. The levels of surface TLR4 showed correlation with the transcription of IL-6 and GP130. TAK242 (TLR4 inhibitor) and PS341 (proteasome inhibitor) can restore the decreased transcription of GP130, both of which were able to promote NF-κB pathway activation in MP-infected cells. These suggested that the regulation of TLR4/NF-κB pathway and induced apoptosis post MP infection are involved in the down-regulation of GP130 at transcription and protein levels, respectively.
Collapse
Affiliation(s)
- Zhikun Zhang
- Department of Pathogenic Biology, School of Basic Medicine Southwest Medical University, Xianglin Road 1#, Luzhou, 646000, China
| | - Dawei Shi
- Tropical Medicine Research Institute, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong-an Road, Xicheng District, Beijing, 100050, China
| | - Haiwei Dou
- Tropical Medicine Research Institute, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong-an Road, Xicheng District, Beijing, 100050, China
| | - Ruijie Wan
- Tropical Medicine Research Institute, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong-an Road, Xicheng District, Beijing, 100050, China
| | - Qing Yuan
- Tropical Medicine Research Institute, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong-an Road, Xicheng District, Beijing, 100050, China
| | - Peng Tu
- Tropical Medicine Research Institute, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong-an Road, Xicheng District, Beijing, 100050, China
| | - Deli Xin
- Tropical Medicine Research Institute, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong-an Road, Xicheng District, Beijing, 100050, China.
| |
Collapse
|
15
|
Osman EEA, Neamati N. Ironing Out the Mechanism of gp130 Signaling. Pharmacol Rev 2024; 76:1399-1443. [PMID: 39414364 DOI: 10.1124/pharmrev.124.001245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 10/18/2024] Open
Abstract
gp130 functions as a shared signal-transducing subunit not only for interleukin (IL)-6 but also for eight other human cytokine receptor complexes. The IL-6 signaling pathway mediated through gp130 encompasses classical, trans, or cluster signaling, intricately regulated by a diverse array of modulators affecting IL-6, its receptor, and gp130. Currently, only a limited number of small molecule antagonists and agonists for gp130 are known. This review aims to comprehensively examine the current knowledge of these modulators and provide insights into their pharmacological properties, particularly in the context of cancer and other diseases. Notably, the prominent gp130 modulators SC144, bazedoxifene, and raloxifene are discussed in detail, with a specific focus on the discovery of SC144's iron-chelating properties. This adds a new dimension to the understanding of its pharmacological effects and therapeutic potential in conditions where iron homeostasis is significant. Our bioinformatic analysis of gp130 and genes related to iron homeostasis reveals insightful correlations, implicating the role of iron in the gp130 signaling pathway. Overall, this review contributes to the evolving understanding of gp130 modulation and its potential therapeutic applications in various disease contexts. SIGNIFICANCE STATEMENT: This perspective provides a timely and comprehensive analysis of advancements in gp130 signaling research, emphasizing the therapeutic implications of the currently available modulators. Bioinformatic analysis demonstrates potential interplay between gp130 and genes that regulate iron homeostasis, suggesting new therapeutic avenues. By combining original research findings with a broader discussion of gp130's therapeutic potential, this perspective significantly contributes to the field.
Collapse
Affiliation(s)
- Essam Eldin A Osman
- Department of Medicinal Chemistry, College of Pharmacy, and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan (E.E.A.O., N.N.) and Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt (E.E.A.O.)
| | - Nouri Neamati
- Department of Medicinal Chemistry, College of Pharmacy, and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan (E.E.A.O., N.N.) and Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt (E.E.A.O.)
| |
Collapse
|
16
|
Divi SN, Markova DZ, D'Antonio ND, Lambrechts MJ, Levy HA, Heard JC, Yalla GR, Chang M, Hilibrand AS, Vaccaro AR, Kepler CK. Circulating microRNAs May Be Predictive of Degenerative Cervical Myelopathy. Spine (Phila Pa 1976) 2024; 49:1393-1400. [PMID: 38711175 DOI: 10.1097/brs.0000000000005025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 04/04/2024] [Indexed: 05/08/2024]
Abstract
STUDY DESIGN Basic Science. OBJECTIVE The objective of this study was to identify a unique serum profile of circulating miRNAs and inflammatory markers in patients with degenerative cervical myelopathy (DCM) compared with healthy controls (HC). SUMMARY OF BACKGROUND DATA Currently, DCM is diagnosed with a combination of history, physical examination, and close correlation to advanced imaging. To date, no serum marker has been identified to be diagnostic of this condition. METHODS Whole venous blood was collected from patients with DCM as well as healthy age-matched and gender-matched controls. miRNA was extracted from venous blood, and a screening analysis was initially conducted to identify miRNA dysregulation in DCM patients. RT-qPCR was used to analyze the expression of 2 specific miRNAs based on screening analysis and literature review. Bioinformatics analysis was used to identify gene networks and potential targets of the miRNA. In addition, the serum inflammatory profile of DCM and HC groups was differentiated using a pro-inflammatory panel. RESULTS Thirty-six patients were enrolled in the DCM group (36.1% male, 61.5±9.5 y), while 35 patients were enrolled in the HC group (31.4% male, 57.5±8.9 y). Of the 15 total miRNAs differentially expressed between DCM and HC groups, two were selected for further analysis: miR-223-3p (upregulated) and miR-451a (downregulated). Functional gene network analysis revealed the highest-ranking gene network was involved in neurological disease, while the most overexpressed miRNA in this network (miR-233-3p) was noted to have over 100 targets, including CDKN1B and the insulin receptor. Serum cytokine analysis showed significant upregulation of several pro-inflammatory cytokines in the DCM cohort compared with the HC group. CONCLUSION DCM patients demonstrated a set of unique circulating miRNAs in addition to a different serum inflammatory profile compared with HC. These miRNAs may potentially serve as targets for future therapeutic intervention or diagnostic/prognostic testing.
Collapse
Affiliation(s)
- Srikanth N Divi
- Department of Orthopaedic Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Dessislava Z Markova
- Department of Orthopaedic Surgery, Sidney Kimmel School of Medicine, Thomas Jefferson University, Philadelphia, PA
| | - Nicholas D D'Antonio
- Department of Orthopaedic Surgery, Sidney Kimmel School of Medicine, Thomas Jefferson University, Philadelphia, PA
| | - Mark J Lambrechts
- Department of Orthopaedic Surgery, Sidney Kimmel School of Medicine, Thomas Jefferson University, Philadelphia, PA
- Rothman Institute, Philadelphia, PA
| | - Hannah A Levy
- Department of Orthopaedic Surgery, Sidney Kimmel School of Medicine, Thomas Jefferson University, Philadelphia, PA
| | - Jeremy C Heard
- Department of Orthopaedic Surgery, Sidney Kimmel School of Medicine, Thomas Jefferson University, Philadelphia, PA
| | - Goutham R Yalla
- Department of Orthopaedic Surgery, Sidney Kimmel School of Medicine, Thomas Jefferson University, Philadelphia, PA
| | - Michael Chang
- Department of Orthopaedic Surgery, Sidney Kimmel School of Medicine, Thomas Jefferson University, Philadelphia, PA
- Rothman Institute, Philadelphia, PA
| | - Alan S Hilibrand
- Department of Orthopaedic Surgery, Sidney Kimmel School of Medicine, Thomas Jefferson University, Philadelphia, PA
- Rothman Institute, Philadelphia, PA
| | - Alexander R Vaccaro
- Department of Orthopaedic Surgery, Sidney Kimmel School of Medicine, Thomas Jefferson University, Philadelphia, PA
- Rothman Institute, Philadelphia, PA
| | - Christopher K Kepler
- Department of Orthopaedic Surgery, Sidney Kimmel School of Medicine, Thomas Jefferson University, Philadelphia, PA
- Rothman Institute, Philadelphia, PA
| |
Collapse
|
17
|
Castorina A, Scheller J, Keay KA, Marzagalli R, Rose-John S, Campbell IL. Increased Expression of the Neuropeptides PACAP/VIP in the Brain of Mice with CNS Targeted Production of IL-6 Is Mediated in Part by Trans-Signalling. Int J Mol Sci 2024; 25:9453. [PMID: 39273398 PMCID: PMC11395455 DOI: 10.3390/ijms25179453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 08/29/2024] [Accepted: 08/29/2024] [Indexed: 09/15/2024] Open
Abstract
Inflammation with expression of interleukin 6 (IL-6) in the central nervous system (CNS) occurs in several neurodegenerative/neuroinflammatory conditions and may cause neurochemical changes to endogenous neuroprotective systems. Pituitary adenylate cyclase-activating polypeptide (PACAP) and vasoactive intestinal polypeptide (VIP) are two neuropeptides with well-established protective and anti-inflammatory properties. Yet, whether PACAP and VIP levels are altered in mice with CNS-restricted, astrocyte-targeted production of IL-6 (GFAP-IL6) remains unknown. In this study, PACAP/VIP levels were assessed in the brain of GFAP-IL6 mice. In addition, we utilised bi-genic GFAP-IL6 mice carrying the human sgp130-Fc transgene (termed GFAP-IL6/sgp130Fc mice) to determine whether trans-signalling inhibition rescued PACAP/VIP changes in the CNS. Transcripts and protein levels of PACAP and VIP, as well as their receptors PAC1, VPAC1 and VPAC2, were significantly increased in the cerebrum and cerebellum of GFAP-IL6 mice vs. wild type (WT) littermates. These results were paralleled by a robust activation of the JAK/STAT3, NF-κB and ERK1/2MAPK pathways in GFAP-IL6 mice. In contrast, co-expression of sgp130Fc in GFAP-IL6/sgp130Fc mice reduced VIP expression and activation of STAT3 and NF-κB pathways, but it failed to rescue PACAP, PACAP/VIP receptors and Erk1/2MAPK phosphorylation. We conclude that forced expression of IL-6 in astrocytes induces the activation of the PACAP/VIP neuropeptide system in the brain, which is only partly modulated upon IL-6 trans-signalling inhibition. Increased expression of PACAP/VIP neuropeptides and receptors may represent a homeostatic response of the CNS to an uncontrolled IL-6 synthesis and its neuroinflammatory consequences.
Collapse
Affiliation(s)
- Alessandro Castorina
- Laboratory of Cellular and Molecular Neuroscience, School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia;
| | - Jurgen Scheller
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine University, 40225 Düsseldorf, Germany;
| | - Kevin A. Keay
- Discipline of Anatomy and Histology, School of Medical Sciences, The University of Sydney, Sydney, NSW 2006, Australia;
| | - Rubina Marzagalli
- Laboratory of Cellular and Molecular Neuroscience, School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia;
| | - Stefan Rose-John
- Institute of Biochemistry, Medical Faculty, Christian Albrechts University, 24098 Kiel, Germany;
| | - Iain L. Campbell
- School of Molecular Bioscience, University of Sydney, Sydney, NSW 2006, Australia;
| |
Collapse
|
18
|
Salar A, Vuković Đerfi K, Pačić A, Škrtić A, Cacev T, Kapitanović S. Association of Functional Polymorphisms in MSH3 and IL-6 Pathway Genes with Different Types of Microsatellite Instability in Sporadic Colorectal Cancer. Cancers (Basel) 2024; 16:2916. [PMID: 39199686 PMCID: PMC11353200 DOI: 10.3390/cancers16162916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 09/01/2024] Open
Abstract
Microsatellite instability (MSI) has been recognized as an important factor in colorectal cancer (CRC). It arises due to deficient mismatch repair (MMR), mostly attributed to MLH1 and MSH2 loss of function leading to a global MMR defect affecting mononucleotide and longer microsatellite loci. Recently, microsatellite instability at tetranucleotide loci, independent of the global MMR defect context, has been suggested to represent a distinct entity with possibly different consequences for tumorigenesis. It arises as a result of an isolated MSH3 loss of function due to its translocation from the nucleus to the cytoplasm under the influence of interleukin-6 (IL-6). In this study the influence of MSH3 and IL-6 signaling pathway polymorphisms (MSH3 exon 1, MSH3+3133A/G, IL-6-174G/C, IL-6R+48892A/C, and gp130+148G/C) on the occurrence of different types of microsatellite instability in sporadic CRC was examined by PCR-RFLP and real-time PCR SNP analyses. A significant difference in distribution of gp130+148G/C genotypes (p = 0.037) and alleles (p = 0.031) was observed in CRC patients with the C allele being less common in tumors with di- and tetranucleotide instability (isolated MSH3 loss of function) compared to tumors without microsatellite instability. A functional polymorphism in gp130 might modulate the IL-6 signaling pathway, directing it toward the occurrence of microsatellite instability corresponding to the IL-6-mediated MSH3 loss of function.
Collapse
Affiliation(s)
- Anamarija Salar
- Laboratory for Personalized Medicine, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (A.S.); (K.V.Đ.)
| | - Kristina Vuković Đerfi
- Laboratory for Personalized Medicine, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (A.S.); (K.V.Đ.)
| | - Arijana Pačić
- Department of Pathology and Cytology, University Hospital Dubrava, 10000 Zagreb, Croatia;
| | - Anita Škrtić
- Department of Pathology and Cytology, University Hospital Merkur, 10000 Zagreb, Croatia;
| | - Tamara Cacev
- Laboratory for Personalized Medicine, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (A.S.); (K.V.Đ.)
| | - Sanja Kapitanović
- Laboratory for Personalized Medicine, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (A.S.); (K.V.Đ.)
| |
Collapse
|
19
|
Bharti V, Kumar A, Wang Y, Roychowdhury N, de Lima Bellan D, Kassaye BB, Watkins R, Capece M, Chung CG, Hilinski G, Vilgelm AE. TTK inhibitor OSU13 promotes immunotherapy responses by activating tumor STING. JCI Insight 2024; 9:e177523. [PMID: 38900577 PMCID: PMC11383830 DOI: 10.1172/jci.insight.177523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 06/18/2024] [Indexed: 06/22/2024] Open
Abstract
TTK spindle assembly checkpoint kinase is an emerging cancer target. This preclinical study explored the antitumor mechanism of TTK inhibitor OSU13 to define a strategy for clinical development. We observed prominent antitumor activity of OSU13 in melanoma, colon and breast cancer cells, organoids derived from patients with melanoma, and mice bearing colon tumors associated with G2 cell cycle arrest, senescence, and apoptosis. OSU13-treated cells displayed DNA damage and micronuclei that triggered the cytosolic DNA-sensing cGAS/STING pathway. STING was required for the induction of several proteins involved in T cell recruitment and activity. Tumors from OSU13-treated mice showed an increased proportion of T and NK cells and evidence of PD-1/PD-L1 immune checkpoint activation. Combining a low-toxicity dose of OSU13 with anti-PD-1 checkpoint blockade resulted in prominent STING- and CD8+ T cell-dependent tumor inhibition and improved survival. These findings provide a rationale for utilizing TTK inhibitors in combination with immunotherapy in STING-proficient tumors.
Collapse
Affiliation(s)
- Vijaya Bharti
- Department of Pathology
- Pelotonia Institute for Immunooncology, and
- Comprehensive Cancer Center, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, Ohio, USA
| | - Amrendra Kumar
- Department of Pathology
- Pelotonia Institute for Immunooncology, and
- Comprehensive Cancer Center, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, Ohio, USA
| | - Yinchong Wang
- Department of Pathology
- Pelotonia Institute for Immunooncology, and
- Comprehensive Cancer Center, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, Ohio, USA
- Molecular Cellular and Developmental Biology Graduate Program, The Ohio State University, Columbus, Ohio, USA
| | - Nikhil Roychowdhury
- Department of Pathology
- Pelotonia Institute for Immunooncology, and
- Comprehensive Cancer Center, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, Ohio, USA
| | - Daniel de Lima Bellan
- Department of Pathology
- Pelotonia Institute for Immunooncology, and
- Comprehensive Cancer Center, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, Ohio, USA
| | | | - Reese Watkins
- Department of Pathology
- Pelotonia Institute for Immunooncology, and
- Comprehensive Cancer Center, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, Ohio, USA
| | - Marina Capece
- Department of Pathology
- Pelotonia Institute for Immunooncology, and
- Comprehensive Cancer Center, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, Ohio, USA
| | | | - Gerard Hilinski
- Drug Development Institute, Comprehensive Cancer Center and The James Cancer Hospital and Solove Research Institute, Columbus, Ohio, USA
| | - Anna E Vilgelm
- Department of Pathology
- Pelotonia Institute for Immunooncology, and
- Comprehensive Cancer Center, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
20
|
Gober IG, Russell AL, Shick TJ, Vagni VA, Carlson JC, Kochanek PM, Wagner AK. Exploratory assessment of the effect of systemic administration of soluble glycoprotein 130 on cognitive performance and chemokine levels in a mouse model of experimental traumatic brain injury. J Neuroinflammation 2024; 21:149. [PMID: 38840141 PMCID: PMC11155101 DOI: 10.1186/s12974-024-03129-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/12/2024] [Indexed: 06/07/2024] Open
Abstract
Uncontrolled neuroinflammation mediates traumatic brain injury (TBI) pathology and impairs recovery. Interleukin-6 (IL-6), a pleiotropic inflammatory regulator, is associated with poor clinical TBI outcomes. IL-6 operates via classical-signaling through membrane-bound IL-6 receptor (IL-6R) and trans-signaling through soluble IL-6 receptor (s)IL-6R. IL-6 trans-signaling specifically contributes to neuropathology, making it a potential precision therapeutic TBI target. Soluble glycoprotein 130 (sgp130) prevents IL-6 trans-signaling, sparing classical signaling, thus is a possible treatment. Mice received either controlled cortical impact (CCI) (6.0 ± 0.2 m/s; 2 mm; 50-60ms) or sham procedures. Vehicle (VEH) or sgp130-Fc was subcutaneously administered to sham (VEH or 1 µg) and CCI (VEH, 0.25 µg or 1 µg) mice on days 1, 4, 7, 10 and 13 post-surgery to assess effects on cognition [Morris Water Maze (MWM)] and ipsilateral hemisphere IL-6 related biomarkers (day 21 post-surgery). CCI + sgp130-Fc groups (0.25 µg and 1 µg) were combined for analysis given similar behavior/biomarker outcomes. CCI + VEH mice had longer latencies and path lengths to the platform and increased peripheral zone time versus Sham + VEH and Sham + sgp130-Fc mice, suggesting injury-induced impairments in learning and anxiety. CCI + sgp130-Fc mice had shorter platform latencies and path lengths and had decreased peripheral zone time, indicating a therapeutic benefit of sgp130-Fc after injury on learning and anxiety. Interestingly, Sham + sgp130-Fc mice had shorter platform latencies, path lengths and peripheral zone times than Sham + VEH mice, suggesting a beneficial effect of sgp130-Fc, independent of injury. CCI + VEH mice had increased brain IL-6 and decreased sgp130 levels versus Sham + VEH and Sham + sgp130-Fc mice. There was no treatment effect on IL-6, sIL6-R or sgp130 in Sham + VEH versus Sham + sgp130-Fc mice. There was also no treatment effect on IL-6 in CCI + VEH versus CCI + sgp130-Fc mice. However, CCI + sgp130-Fc mice had increased sIL-6R and sgp130 versus CCI + VEH mice, demonstrating sgp130-Fc treatment effects on brain biomarkers. Inflammatory chemokines (MIP-1β, IP-10, MIG) were increased in CCI + VEH mice versus Sham + VEH and Sham + sgp130-Fc mice. However, CCI + sgp130-Fc mice had decreased chemokine levels versus CCI + VEH mice. IL-6 positively correlated, while sgp130 negatively correlated, with chemokine levels. Overall, we found that systemic sgp130-Fc treatment after CCI improved learning, decreased anxiety and reduced CCI-induced brain chemokines. Future studies will explore sex-specific dosing and treatment mechanisms for sgp130-Fc therapy.
Collapse
Affiliation(s)
- Ian G Gober
- Department of Physical Medicine and Rehabilitation, School of Medicine, University of Pittsburgh, 3471 Fifth Avenue, Suite 910, Pittsburgh, PA, 15213, USA
- Safar Center for Resuscitation Research, John G. Rangos Research Center, Pittsburgh, PA, USA
| | - Ashley L Russell
- Department of Physical Medicine and Rehabilitation, School of Medicine, University of Pittsburgh, 3471 Fifth Avenue, Suite 910, Pittsburgh, PA, 15213, USA
- Safar Center for Resuscitation Research, John G. Rangos Research Center, Pittsburgh, PA, USA
| | - Tyler J Shick
- Department of Physical Medicine and Rehabilitation, School of Medicine, University of Pittsburgh, 3471 Fifth Avenue, Suite 910, Pittsburgh, PA, 15213, USA
- Safar Center for Resuscitation Research, John G. Rangos Research Center, Pittsburgh, PA, USA
| | - Vincent A Vagni
- Safar Center for Resuscitation Research, John G. Rangos Research Center, Pittsburgh, PA, USA
- Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jenna C Carlson
- Department of Biostatistics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Patrick M Kochanek
- Safar Center for Resuscitation Research, John G. Rangos Research Center, Pittsburgh, PA, USA
- Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Amy K Wagner
- Department of Physical Medicine and Rehabilitation, School of Medicine, University of Pittsburgh, 3471 Fifth Avenue, Suite 910, Pittsburgh, PA, 15213, USA.
- Safar Center for Resuscitation Research, John G. Rangos Research Center, Pittsburgh, PA, USA.
- Center for Neuroscience, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Neuroscience, School of Arts and Sciences, University of Pittsburgh, Pittsburgh, PA, USA.
- Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
21
|
Xu S, Deng KQ, Lu C, Fu X, Zhu Q, Wan S, Zhang L, Huang Y, Nie L, Cai H, Wang Q, Zeng H, Zhang Y, Wang F, Ren H, Chen Y, Yan H, Xu K, Zhou L, Lu M, Zhu Y, Liu S, Lu Z. Interleukin-6 classic and trans-signaling utilize glucose metabolism reprogramming to achieve anti- or pro-inflammatory effects. Metabolism 2024; 155:155832. [PMID: 38438106 DOI: 10.1016/j.metabol.2024.155832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 02/26/2024] [Accepted: 02/29/2024] [Indexed: 03/06/2024]
Abstract
Interleukin (IL)-6 has anti- and pro-inflammatory functions, controlled by IL-6 classic and trans-signaling, respectively. Differences in the downstream signaling mechanism between IL-6 classic and trans-signaling have not been identified. Here, we report that IL-6 activates glycolysis to regulate the inflammatory response. IL-6 regulates glucose metabolism by forming a complex containing signal-transducing activators of transcription 3 (STAT3), hexokinase 2 (HK2), and voltage-dependent anion channel 1 (VDAC1). The IL-6 classic signaling directs glucose flux to oxidative phosphorylation (OxPhos), while IL-6 trans-signaling directs glucose flux to anaerobic glycolysis. Classic IL-6 signaling promotes STAT3 translocation into mitochondria to interact with pyruvate dehydrogenase kinase-1 (PDK1), leading to pyruvate dehydrogenase α (PDHA) dissociation from PDK1. As a result, PDHA is dephosphorylated, and STAT3 is phosphorylated at Ser727. By contrast, IL-6 trans-signaling promotes the interaction of sirtuin 2 (SIRT2) and lactate dehydrogenase A (LDHA), leading to the dissociation of STAT3 from SIRT2. As a result, LDHA is deacetylated, and STAT3 is acetylated and phosphorylated at Tyr705. IL-6 classic signaling promotes the differentiation of regulatory T cells via the PDK1/STAT3/PDHA axis, whereas IL-6 trans-signaling promotes the differentiation of Th17 cells via the SIRT2/STAT3/LDHA axis. Conclusion: IL-6 classic signaling generates anti-inflammatory functions by shifting energy metabolism to OxPhos, while IL-6 trans-signaling generates pro-inflammatory functions by shifting energy metabolism to anaerobic glycolysis.
Collapse
Affiliation(s)
- Shilei Xu
- Department of General Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510530, China.
| | - Ke-Qiong Deng
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan 430072, China; Institute of Myocardial Injury and Repair, Wuhan University, Wuhan 430072, China.
| | - Chengbo Lu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Taikang Center for Life and Medical Sciences, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430072, China
| | - Xin Fu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Taikang Center for Life and Medical Sciences, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430072, China
| | - Qingmei Zhu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Taikang Center for Life and Medical Sciences, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430072, China.
| | - Shiqi Wan
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Taikang Center for Life and Medical Sciences, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430072, China.
| | - Lin Zhang
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan 430072, China; Institute of Myocardial Injury and Repair, Wuhan University, Wuhan 430072, China
| | - Yu Huang
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan 430072, China.
| | - Longyu Nie
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan 430072, China.
| | - Huanhuan Cai
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan 430072, China; Institute of Myocardial Injury and Repair, Wuhan University, Wuhan 430072, China.
| | - Qiming Wang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, Human Province, China
| | - Hao Zeng
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, China.
| | - Yufeng Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, China.
| | - Fubing Wang
- Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan 430072, China
| | - Hong Ren
- Shanghai Children's Medical Center, Affiliated Hospital to Shanghai Jiao Tong University School of Medicine, China.
| | - Yu Chen
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Taikang Center for Life and Medical Sciences, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430072, China.
| | - Huan Yan
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Taikang Center for Life and Medical Sciences, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430072, China.
| | - Ke Xu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Taikang Center for Life and Medical Sciences, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430072, China.
| | - Li Zhou
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Taikang Center for Life and Medical Sciences, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430072, China.
| | - Mengji Lu
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen 45122, Germany.
| | - Ying Zhu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Taikang Center for Life and Medical Sciences, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430072, China.
| | - Shi Liu
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan 430072, China; Institute of Myocardial Injury and Repair, Wuhan University, Wuhan 430072, China; State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Taikang Center for Life and Medical Sciences, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430072, China; College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, Human Province, China.
| | - Zhibing Lu
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan 430072, China; Institute of Myocardial Injury and Repair, Wuhan University, Wuhan 430072, China.
| |
Collapse
|
22
|
Rodríguez-Hernández MÁ, Baena-Bustos M, Carneros D, Zurita-Palomo C, Muñoz-Pinillos P, Millán J, Padillo FJ, Smerdou C, von Kobbe C, Rose-John S, Bustos M. Targeting IL-6 trans-signalling by sgp130Fc attenuates severity in SARS-CoV-2 -infected mice and reduces endotheliopathy. EBioMedicine 2024; 103:105132. [PMID: 38677182 PMCID: PMC11061249 DOI: 10.1016/j.ebiom.2024.105132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 04/02/2024] [Accepted: 04/08/2024] [Indexed: 04/29/2024] Open
Abstract
BACKGROUND SARS-CoV-2 infection is considered as a relapsing inflammatory process with a dysregulation of IL-6 signalling. Classic IL-6 signalling is thought to represent a defence mechanism against pathogens. In contrast, IL-6 trans-signalling has pro-inflammatory effects. In severe COVID-19, therapeutic strategies have focused on global inhibition of IL-6, with controversial results. We hypothesized that specific blockade of IL-6 trans-signalling could inhibit inflammatory response preserving the host defence activity inherent to IL-6 classic signalling. METHODS To test the role of the specific IL-6 trans-signalling inhibition by sgp130Fc in short- and long-term consequences of COVID-19, we used the established K18-hACE2 transgenic mouse model. Histological as well as immunohistochemical analysis, and pro-inflammatory marker profiling were performed. To investigate IL-6 trans-signalling in human cells we used primary lung microvascular endothelial cells and fibroblasts in the presence/absence of sgp130Fc. FINDINGS We report that targeting IL-6 trans-signalling by sgp130Fc attenuated SARS-CoV-2-related clinical symptoms and mortality. In surviving mice, the treatment caused a significant decrease in lung damage. In vitro, IL-6 trans-signalling induced strong and persisting JAK1/STAT3 activation in endothelial cells and lung fibroblasts with proinflammatory effects, which were attenuated by sgp130Fc. Our data also suggest that in those cells with scant amounts of IL-6R, the induction of gp130 and IL-6 by IL-6:sIL-6R complex sustains IL-6 trans-signalling. INTERPRETATION IL-6 trans-signalling fosters progression of COVID-19, and suggests that specific blockade of this signalling mode could offer a promising alternative to mitigate both short- and long-term consequences without affecting the beneficial effects of IL-6 classic signalling. These results have implications for the development of new therapies of lung injury and endotheliopathy in COVID-19. FUNDING The project was supported by ISCIII, Spain (COV-20/00792 to MB, PI23/01351 to MARH) and the European Commission-Next generation EU (European Union) (Regulation EU 2020/2094), through CSIC's Global Health Platform (PTI Salud Global, SGL2103029 to MB). PID2019-110587RB-I00 (MB) supported by MICIN/AEI/10.13039/501100011033/and PID2022-143034OB-I00 (MB) by MICIN/AEI/10.13039/501100011033/FEDER. MAR-H acknowledges support from ISCIII, Spain and the European Commission-Next generation EU (European Union), through CSIC's Global Health PTI.
Collapse
Affiliation(s)
- María Ángeles Rodríguez-Hernández
- Area of Liver, Digestive and Inflammatory Diseases, Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital (HUVR), Spanish National Research Council (CSIC), University of Seville (US), Seville, Spain.
| | - Mercedes Baena-Bustos
- Pneumology Unit, Institute of Biomedicine of Seville (IBiS), Virgen Macarena University Hospital (HUVM), Spanish National Research Council (CSIC), University of Seville (US), Seville, Spain
| | - David Carneros
- Area of Liver, Digestive and Inflammatory Diseases, Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital (HUVR), Spanish National Research Council (CSIC), University of Seville (US), Seville, Spain
| | - Carola Zurita-Palomo
- Area of Liver, Digestive and Inflammatory Diseases, Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital (HUVR), Spanish National Research Council (CSIC), University of Seville (US), Seville, Spain
| | - Pablo Muñoz-Pinillos
- Centro de Biología Molecular Severo Ochoa (CBM), CSIC-UAM, Cantoblanco, Madrid, Spain
| | - Jaime Millán
- Centro de Biología Molecular Severo Ochoa (CBM), CSIC-UAM, Cantoblanco, Madrid, Spain
| | - Francisco Javier Padillo
- Area of Liver, Digestive and Inflammatory Diseases, Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital (HUVR), Spanish National Research Council (CSIC), University of Seville (US), Seville, Spain
| | - Cristian Smerdou
- Division of DNA and RNA Medicine, Cima Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IdISNA), and CCUN, Pamplona, Spain
| | - Cayetano von Kobbe
- Centro de Biología Molecular Severo Ochoa (CBM), CSIC-UAM, Cantoblanco, Madrid, Spain
| | | | - Matilde Bustos
- Area of Liver, Digestive and Inflammatory Diseases, Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital (HUVR), Spanish National Research Council (CSIC), University of Seville (US), Seville, Spain.
| |
Collapse
|
23
|
Rafii P, Cruz PR, Ettich J, Seibel C, Padrini G, Wittich C, Lang A, Petzsch P, Köhrer K, Moll JM, Floss DM, Scheller J. Engineered interleukin-6-derived cytokines recruit artificial receptor complexes and disclose CNTF signaling via the OSMR. J Biol Chem 2024; 300:107251. [PMID: 38569939 PMCID: PMC11039321 DOI: 10.1016/j.jbc.2024.107251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 03/22/2024] [Accepted: 03/27/2024] [Indexed: 04/05/2024] Open
Abstract
Ciliary neurotrophic factor (CNTF) activates cells via the non-signaling α-receptor CNTF receptor (CNTFR) and the two signaling β-receptors glycoprotein 130 (gp130) and leukemia inhibitory factor receptor (LIFR). The CNTF derivate, Axokine, was protective against obesity and insulin resistance, but clinical development was halted by the emergence of CNTF antibodies. The chimeric cytokine IC7 used the framework of interleukin (IL-)6 with the LIFR-binding site from CNTF to activate cells via IL-6R:gp130:LIFR complexes. Similar to CNTF/Axokine, IC7 protected mice from obesity and insulin resistance. Here, we developed CNTF-independent chimeras that specifically target the IL-6R:gp130:LIFR complex. In GIL-6 and GIO-6, we transferred the LIFR binding site from LIF or OSM to IL-6, respectively. While GIO-6 signals via gp130:IL-6R:LIFR and gp130:IL-6R:OSMR complexes, GIL-6 selectively activates the IL-6R:gp130:LIFR receptor complex. By re-evaluation of IC7 and CNTF, we discovered the Oncostatin M receptor (OSMR) as an alternative non-canonical high-affinity receptor leading to IL-6R:OSMR:gp130 and CNTFR:OSMR:gp130 receptor complexes, respectively. The discovery of OSMR as an alternative high-affinity receptor for IC7 and CNTF designates GIL-6 as the first truly selective IL-6R:gp130:LIFR cytokine, whereas GIO-6 is a CNTF-free alternative for IC7.
Collapse
Affiliation(s)
- Puyan Rafii
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Patricia Rodrigues Cruz
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Julia Ettich
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Christiane Seibel
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Giacomo Padrini
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Christoph Wittich
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Alexander Lang
- Division of Cardiology, Pulmonology, and Vascular Medicine, Cardiovascular Research Laboratory, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Patrick Petzsch
- Biological and Medical Research Center (BMFZ), Medical Faculty, Heinrich-Heine-University, Duesseldorf, Germany
| | - Karl Köhrer
- Biological and Medical Research Center (BMFZ), Medical Faculty, Heinrich-Heine-University, Duesseldorf, Germany
| | - Jens M Moll
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Doreen M Floss
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Jürgen Scheller
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany.
| |
Collapse
|
24
|
Scheller J, Ettich J, Wittich C, Pudewell S, Floss DM, Rafii P. Exploring the landscape of synthetic IL-6-type cytokines. FEBS J 2024; 291:2030-2050. [PMID: 37467060 DOI: 10.1111/febs.16909] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/30/2023] [Accepted: 07/17/2023] [Indexed: 07/21/2023]
Abstract
Interleukin-6 (IL-6)-type cytokines not only have key immunomodulatory functions that affect the pathogenesis of diseases such as autoimmune diseases, chronic inflammatory conditions, and cancer, but also fulfill important homeostatic tasks. Even though the pro-inflammatory arm has hindered the development of therapeutics based on natural-like IL-6-type cytokines to date, current synthetic trends might pave the way to overcome these limitations and eventually lead to immune-inert designer cytokines to aid type 2 diabetes and brain injuries. Those synthetic biology approaches include mutations, fusion proteins, and inter-cytokine swapping, and resulted in IL-6-type cytokines with altered receptor affinities, extended target cell profiles, and targeting of non-natural cytokine receptor complexes. Here, we survey synthetic cytokine developments within the IL-6-type cytokine family and discuss potential clinical applications.
Collapse
Affiliation(s)
- Jürgen Scheller
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Julia Ettich
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Christoph Wittich
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Silke Pudewell
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Doreen M Floss
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Puyan Rafii
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| |
Collapse
|
25
|
Korotaeva AA, Samoilova EV, Pogosova NV, Kuchiev DT, Gomyranova NV, Paleev FN. Factors of Interleukin-6 Signaling in COVID-19 Patients with Lung Damage of Varying Degrees: A Pilot Study. Bull Exp Biol Med 2024; 176:772-775. [PMID: 38890212 DOI: 10.1007/s10517-024-06106-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Indexed: 06/20/2024]
Abstract
Specific features of IL-6 signal transduction were studied in 89 patients with lung damage of varying degrees during the first COVID-19 pandemic wave. The levels of IL-6 signaling components (IL-6, sIL-6R, and sgp130) and highly sensitive C-reactive protein (hsCRP) were examined in patients with intact lungs (CT-0), mild (CT-1), moderate (CT-2), moderate to severe (CT-3), and severe (CT-4) lung damage. Seventy patients were re-examined 3-7 months after discharge from the hospital. The IL-6 and hsCRP levels increased several times with severing lung damage severity. In patients with CT-3, sIL6-R increased statistically significantly and remained high in CT-4 patients. sgp130 levels were lower in CT-1 and CT-2 patients and higher in CT-3 and CT-4 patients compared to CT-0 patients. We revealed a positive correlation between IL-6 and hsCRP levels in CT-1, CT-2, and CT-3 patients. In CT-3 patients, sIL-6R levels positively correlated with IL-6 concentration. The studied parameters decreased considerably in all patients 3-7 months after discharge. It can be suggested that IL-6 classic-signaling is predominant in CT-1 and CT-2, while trans-signaling prevails in CT-3. Disorders in regulatory mechanisms of IL-6 signaling occur in CT-4, which prevents physiological elimination of IL-6 hyperactivity. The results obtained are preliminary and require a broader study.
Collapse
Affiliation(s)
- A A Korotaeva
- E. I. Chazov National Medical Research Center of Cardiology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - E V Samoilova
- E. I. Chazov National Medical Research Center of Cardiology, Ministry of Health of the Russian Federation, Moscow, Russia.
| | - N V Pogosova
- E. I. Chazov National Medical Research Center of Cardiology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - D T Kuchiev
- E. I. Chazov National Medical Research Center of Cardiology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - N V Gomyranova
- E. I. Chazov National Medical Research Center of Cardiology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - F N Paleev
- E. I. Chazov National Medical Research Center of Cardiology, Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
26
|
Vlashi R, Zhang X, Li H, Chen G. Potential therapeutic strategies for osteoarthritis via CRISPR/Cas9 mediated gene editing. Rev Endocr Metab Disord 2024; 25:339-367. [PMID: 38055160 DOI: 10.1007/s11154-023-09860-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/28/2023] [Indexed: 12/07/2023]
Abstract
Osteoarthritis (OA) is an incapacitating and one of the most common physically degenerative conditions with an assorted etiology and a highly complicated molecular mechanism that to date lacks an efficient treatment. The capacity to design biological networks and accurately modify existing genomic sites holds an apt potential for applications across medical and biotechnological sciences. One of these highly specific genomes editing technologies is the CRISPR/Cas9 mechanism, referred to as the clustered regularly interspaced short palindromic repeats, which is a defense mechanism constituted by CRISPR associated protein 9 (Cas9) directed by small non-coding RNAs (sncRNA) that bind to target DNA through Watson-Crick base pairing rules where subsequent repair of the target DNA is initiated. Up-to-date research has established the effectiveness of the CRISPR/Cas9 mechanism in targeting the genetic and epigenetic alterations in OA by suppressing or deleting gene expressions and eventually distributing distinctive anti-arthritic properties in both in vitro and in vivo osteoarthritic models. This review aims to epitomize the role of this high-throughput and multiplexed gene editing method as an analogous therapeutic strategy that could greatly facilitate the clinical development of OA-related treatments since it's reportedly an easy, minimally invasive technique, and a comparatively less painful method for osteoarthritic patients.
Collapse
Affiliation(s)
- Rexhina Vlashi
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Xingen Zhang
- Department of Orthopedics, Jiaxing Key Laboratory for Minimally Invasive Surgery in Orthopaedics & Skeletal Regenerative Medicine, Zhejiang Rongjun Hospital, Jiaxing, 314001, China
| | - Haibo Li
- The Central Laboratory of Birth Defects Prevention and Control, Ningbo Women and Children's Hospital, Ningbo, China.
- Ningbo Key Laboratory for the Prevention and Treatment of Embryogenic Diseases, Ningbo Women and Children's Hospital, Ningbo, China.
| | - Guiqian Chen
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China.
| |
Collapse
|
27
|
Cossu AM, Melisi F, Noviello TMR, Pasquale LS, Grisolia P, Reale C, Bocchetti M, Falco M, Tammaro C, Accardo N, Longo F, Allosso S, Mesolella M, Addeo R, Perri F, Ottaiano A, Ricciardiello F, Amler E, Ambrosino C, Misso G, Ceccarelli M, Caraglia M, Scrima M. MiR-449a antagonizes EMT through IL-6-mediated trans-signaling in laryngeal squamous cancer. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102140. [PMID: 38425711 PMCID: PMC10901858 DOI: 10.1016/j.omtn.2024.102140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 02/01/2024] [Indexed: 03/02/2024]
Abstract
MicroRNAs (miRNAs) are involved in post-transcriptional gene expression regulation and in mechanisms of cancer growth and metastases. In this light, miRNAs could be promising therapeutic targets and biomarkers in clinical practice. Therefore, we investigated if specific miRNAs and their target genes contribute to laryngeal squamous cell carcinoma (LSCC) development. We found a significant decrease of miR-449a in LSCC patients with nodal metastases (63.3%) compared with patients without nodal involvement (44%). The AmpliSeq Transcriptome of HNO-210 miR-449a-transfected cell lines allowed the identification of IL6-R as a potential target. Moreover, the downregulation of IL6-R and the phosphorylation reduction of the downstream signaling effectors, suggested the inhibition of the IL-6 trans-signaling pathway. These biochemical effects were paralleled by a significant inhibition of invasion and migration in vitro and in vivo, supporting an involvement of epithelial-mesenchymal transition. These findings indicate that miR-449a contributes to suppress the metastasization of LSCC by the IL-6 trans-signaling block and affects sensitivity to external stimuli that mimic pro-inflammatory conditions.
Collapse
Affiliation(s)
- Alessia Maria Cossu
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
- Biogem Scarl, Institute of Genetic Research, 83031 Ariano Irpino, Italy
| | - Federica Melisi
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
- Biogem Scarl, Institute of Genetic Research, 83031 Ariano Irpino, Italy
| | - Teresa Maria Rosaria Noviello
- Biogem Scarl, Institute of Genetic Research, 83031 Ariano Irpino, Italy
- Department of Electrical Engineering and Information Technology, University of Naples "Federico II", Napoli, Italy
| | - Lucia Stefania Pasquale
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
- Biogem Scarl, Institute of Genetic Research, 83031 Ariano Irpino, Italy
| | - Piera Grisolia
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
- Biogem Scarl, Institute of Genetic Research, 83031 Ariano Irpino, Italy
| | - Carla Reale
- Biogem Scarl, Institute of Genetic Research, 83031 Ariano Irpino, Italy
| | - Marco Bocchetti
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
- Biogem Scarl, Institute of Genetic Research, 83031 Ariano Irpino, Italy
| | - Michela Falco
- Biogem Scarl, Institute of Genetic Research, 83031 Ariano Irpino, Italy
| | - Chiara Tammaro
- Biogem Scarl, Institute of Genetic Research, 83031 Ariano Irpino, Italy
| | - Nunzio Accardo
- Ear, Nose, and Throat Unit, AORN "Antonio Cardarelli", Naples, Italy
| | - Francesco Longo
- Head and Neck Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione G. Pascale", IRCCS, Naples, Italy
| | - Salvatore Allosso
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, UOC Federico II, 80121 Naples, Italy
| | - Massimo Mesolella
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, UOC Federico II, 80121 Naples, Italy
| | - Raffaele Addeo
- Medical Oncology Unit, San Giovanni di Dio Hospital, 80027 Frattamaggiore, Italy
| | - Francesco Perri
- Head and Neck Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione G. Pascale", IRCCS, Naples, Italy
| | - Alessandro Ottaiano
- SSD Innovative Therapies for Abdominal Metastases, Abdominal Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione G. Pascale", IRCCS, Naples, Italy
| | | | - Evzen Amler
- UCEEB, Czech Technical University, Třinecká 1024, 273 43 Buštěhrad, Czech
| | - Concetta Ambrosino
- Biogem Scarl, Institute of Genetic Research, 83031 Ariano Irpino, Italy
- Department of Science and Technology, University of Sannio, 82100 Benevento, Italy
| | - Gabriella Misso
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Michele Ceccarelli
- Biogem Scarl, Institute of Genetic Research, 83031 Ariano Irpino, Italy
- Department of Electrical Engineering and Information Technology, University of Naples "Federico II", Napoli, Italy
| | - Michele Caraglia
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
- Biogem Scarl, Institute of Genetic Research, 83031 Ariano Irpino, Italy
| | - Marianna Scrima
- Biogem Scarl, Institute of Genetic Research, 83031 Ariano Irpino, Italy
| |
Collapse
|
28
|
Park EJ, Lee CW. Soluble receptors in cancer: mechanisms, clinical significance, and therapeutic strategies. Exp Mol Med 2024; 56:100-109. [PMID: 38182653 PMCID: PMC10834419 DOI: 10.1038/s12276-023-01150-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/31/2023] [Accepted: 11/06/2023] [Indexed: 01/07/2024] Open
Abstract
Soluble receptors are soluble forms of receptors found in the extracellular space. They have emerged as pivotal regulators of cellular signaling and disease pathogenesis. This review emphasizes their significance in cancer as diagnostic/prognostic markers and potential therapeutic targets. We provide an overview of the mechanisms by which soluble receptors are generated along with their functions. By exploring their involvement in cancer progression, metastasis, and immune evasion, we highlight the importance of soluble receptors, particularly soluble cytokine receptors and immune checkpoints, in the tumor microenvironment. Although current research has illustrated the emerging clinical relevance of soluble receptors, their therapeutic applications remain underexplored. As the landscape of cancer treatment evolves, understanding and targeting soluble receptors might pave the way for novel strategies for cancer diagnosis, prognosis, and therapy.
Collapse
Affiliation(s)
- Eun-Ji Park
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Chang-Woo Lee
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea.
- SKKU Institute for Convergence, Sungkyunkwan University, Suwon, Republic of Korea.
| |
Collapse
|
29
|
Rodríguez J, De Santis Arévalo J, Dennis VA, Rodríguez AM, Giambartolomei GH. Bystander activation of microglia by Brucella abortus-infected astrocytes induces neuronal death via IL-6 trans-signaling. Front Immunol 2024; 14:1343503. [PMID: 38322014 PMCID: PMC10844513 DOI: 10.3389/fimmu.2023.1343503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 12/29/2023] [Indexed: 02/08/2024] Open
Abstract
Inflammation plays a key role in the pathogenesis of neurobrucellosis where glial cell interactions are at the root of this pathological condition. In this study, we present evidence indicating that soluble factors secreted by Brucella abortus-infected astrocytes activate microglia to induce neuronal death. Culture supernatants (SN) from B. abortus-infected astrocytes induce the release of pro-inflammatory mediators and the increase of the microglial phagocytic capacity, which are two key features in the execution of live neurons by primary phagocytosis, a recently described mechanism whereby B. abortus-activated microglia kills neurons by phagocytosing them. IL-6 neutralization completely abrogates neuronal loss. IL-6 is solely involved in increasing the phagocytic capacity of activated microglia as induced by SN from B. abortus-infected astrocytes and does not participate in their inflammatory activation. Both autocrine microglia-derived and paracrine astrocyte-secreted IL-6 endow microglial cells with up-regulated phagocytic capacity that allows them to phagocytose neurons. Blocking of IL-6 signaling by soluble gp130 abrogates microglial phagocytosis and concomitant neuronal death, indicating that IL-6 activates microglia via trans-signaling. Altogether, these results demonstrate that soluble factors secreted by B. abortus-infected astrocytes activate microglia to induce, via IL-6 trans-signaling, the death of neurons. IL-6 signaling inhibition may thus be considered a strategy to control inflammation and CNS damage in neurobrucellosis.
Collapse
Affiliation(s)
- Julia Rodríguez
- Instituto de Inmunología, Genética y Metabolismo (INIGEM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Julia De Santis Arévalo
- Instituto de Inmunología, Genética y Metabolismo (INIGEM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Vida A Dennis
- Center for NanoBiotechnology Research and Department of Biological Sciences, Alabama State University, Montgomery, AL, United States
| | - Ana M Rodríguez
- Instituto de Inmunología, Genética y Metabolismo (INIGEM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Guillermo H Giambartolomei
- Instituto de Inmunología, Genética y Metabolismo (INIGEM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
30
|
Widjaja AA, Cook SA. Nonspecific Inhibition of IL6 Family Cytokine Signalling by Soluble gp130. Int J Mol Sci 2024; 25:1363. [PMID: 38338642 PMCID: PMC10855816 DOI: 10.3390/ijms25031363] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/17/2024] [Accepted: 01/21/2024] [Indexed: 02/12/2024] Open
Abstract
IL6 is a proinflammatory cytokine that binds to membrane-bound IL6 receptor (IL6R) or soluble IL6R to signal via gp130 in cis or trans, respectively. We tested the hypothesis that sgp130Fc, which is believed to be a selective IL6 trans-signalling inhibitor, is in fact a non-specific inhibitor of gp130 signalling. In human cancer and primary cells, sgp130Fc inhibited IL6, IL11, OSM and CT1 cis-signalling. The IC50 values of sgp130Fc for IL6 and OSM cis-signalling were markedly (20- to 200-fold) lower than the concentrations of sgp130Fc used in mouse studies and clinical trials. sgp130 inhibited IL6 and OSM signalling in the presence of an ADAM10/17 inhibitor and the absence of soluble IL6R or OSMR, with effects that were indistinguishable from those of a gp130 neutralising antibody. These data show that sgp130Fc does not exclusively block IL6 trans-signalling and reveal instead that broad inhibition of gp130 signalling likely underlies its therapeutic effects. This proposes global or modular inhibition of gp130 as a therapeutic approach for treating human disease.
Collapse
Affiliation(s)
- Anissa A. Widjaja
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, Singapore 169857, Singapore
| | - Stuart A. Cook
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, Singapore 169857, Singapore
- National Heart Centre Singapore, National Heart Research Institute Singapore, Singapore 169609, Singapore
- MRC-London Institute of Medical Sciences, Hammersmith Hospital Campus, London W6 8RF, UK
| |
Collapse
|
31
|
Deng W, Zhao Z, Zou T, Kuang T, Wang J. Research Advances in Fusion Protein-Based Drugs for Diabetes Treatment. Diabetes Metab Syndr Obes 2024; 17:343-362. [PMID: 38288338 PMCID: PMC10823413 DOI: 10.2147/dmso.s421527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 12/22/2023] [Indexed: 01/31/2024] Open
Abstract
Diabetes mellitus (DM) is a chronic metabolic disease characterized by elevated blood glucose levels, resulting in multi-organ dysfunction and various complications. Fusion proteins can form multifunctional complexes by combining the target proteins with partner proteins. It has significant advantages in improving the performance of the target proteins, extending their biological half-life, and enhancing patient drug compliance. Fusion protein-based drugs have emerged as promising new drugs in diabetes therapeutics. However, there has not been a systematic review of fusion protein-based drugs for diabetes therapeutics. Hence, we conducted a comprehensive review of published literature on diabetic fusion protein-based drugs for diabetes, with a primary focus on immunoglobulin G (IgG) fragment crystallizable (Fc) region, albumin, and transferrin (TF). This review aims to provide a reference for the subsequent development and clinical application of fusion protein-based drugs in diabetes therapeutics.
Collapse
Affiliation(s)
- Wenying Deng
- School of Basic Medical Sciences, University of South China, Hengyang, Hunan Province, 421001, People’s Republic of China
| | - Zeyi Zhao
- School of Basic Medical Sciences, University of South China, Hengyang, Hunan Province, 421001, People’s Republic of China
| | - Tao Zou
- Department of Cardiovascular Medicine, First Affiliated Hospital of University of South China, Hengyang, Hunan Province, 421001, People’s Republic of China
| | - Tongdong Kuang
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi Province, 541199, People’s Republic of China
| | - Jing Wang
- School of Basic Medical Sciences, University of South China, Hengyang, Hunan Province, 421001, People’s Republic of China
| |
Collapse
|
32
|
Chen YF, Li SC, Huang EY. Role of microbiota in radiation-induced small-bowel damage. JOURNAL OF RADIATION RESEARCH 2024; 65:55-62. [PMID: 37996087 PMCID: PMC10803162 DOI: 10.1093/jrr/rrad084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/24/2023] [Accepted: 10/10/2023] [Indexed: 11/25/2023]
Abstract
Radiation-induced gastrointestinal damage is a common acute radiation syndrome. Previous studies have highlighted that Galectin-1 and Interleukin-6 (IL-6) are associated with flaking of small intestinal villi and intestinal radioresistance. Therefore, our goal is to study whether gut bacteria regulated by galectin-1 or IL-6 can mitigate radiation-induced small intestine damage. In this study, differences between galectin-1, sgp130-regulated and wild-type (WT) mice were analyzed by microbiome array. The effects of the Firmicutes/Bacteroidetes (F/B) ratio and the proportion of bacterial distribution at the phylum level were observed after 18 Gy whole abdomen radiation. Fecal microbiota transplantation was used to implant radioresistant gut flora into WT mice, and the number of viable small intestinal crypt foci was observed by immunohistochemistry. Fecal transplantation from galectin-1 knockout and sgp130 transgenic mice, with higher radiation resistance, into WT mice significantly increased the number of surviving small intestinal crypts. This radiation resistance, generated through gene regulation, was not affected by the F/B ratio. We initially found that the small intestinal villi of WT mice receiving radioresistant mouse fecal bacteria demonstrated better repair outcomes after radiation exposure. These results indicate the need for a focus on the identification and application of superior radioresistant bacterial strains. In our laboratory, we will further investigate specific radioresistant bacterial strains to alleviate acute side effects of radiation therapy to improve the patients' immune ability and postoperative quality of life.
Collapse
Affiliation(s)
- Yi-Fan Chen
- Department of Radiation Oncology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, 129, Da-Pi Road, Niao-Sung District, Kaohsiung 833401, Taiwan
- Department and Graduate Institute of Aquaculture, National Kaohsiung University of Science and Technology, 142, Haizhuan Road, Nanzi District, Kaohsiung 811213, Taiwan
| | - Sung-Chou Li
- Genomics and Proteomics Core Laboratory, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, 129, Da-Pi Road, Niao-Sung District, Kaohsiung 833401, Taiwan
| | - Eng-Yen Huang
- Department of Radiation Oncology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, 129, Da-Pi Road, Niao-Sung District, Kaohsiung 833401, Taiwan
- School of Medicine, College of Medicine, National Sun Yat-sen University, 70, Lienhai Road, Gushan District, Kaohsiung 80424, Taiwan
| |
Collapse
|
33
|
Avci AB, Feist E, Burmester GR. Targeting IL-6 or IL-6 Receptor in Rheumatoid Arthritis: What Have We Learned? BioDrugs 2024; 38:61-71. [PMID: 37989892 PMCID: PMC10789669 DOI: 10.1007/s40259-023-00634-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/18/2023] [Indexed: 11/23/2023]
Abstract
The use of different pathways in the treatment of rheumatoid arthritis has led to a significant decrease in the number of treatment-resistant patients. In this context, interleukin (IL)-6 inhibition has filled an important gap in rheumatoid arthritis treatment with its effectiveness and safety in both monotherapy and combinations. The process of IL-6 inhibition initiated with IL-6 receptor blockers has prompted questions regarding the potential impact and safety of different inhibitions of this pathway, such as the direct blockade of IL-6. Following the termination of the development of sirukumab because of mortality data in early studies, the investigation of olokizumab, which targets a different region of the IL-6 cytokine, has renewed the hope in this area and the safety concerns have been largely alleviated by the open-label extension data. In addition, the efficacy and safety of tocilizumab and sarilumab have led to a rapid investigation of biosimilars and new potent IL-6 receptor blockers. A comprehensive understanding of mechanisms of this pathway with further long-term clinical data and basic research may provide a decisive impact on selecting the appropriate mechanism as the first choice in personalized treatments.
Collapse
Affiliation(s)
- Ali Berkant Avci
- Department of Internal Medicine, Rheumatology, Medical Park Antalya Hospital, Antalya, Türkiye
| | - Eugen Feist
- Department of Rheumatology, Helios Fachklinik Vogelsang-Gommern, Cooperation Partner of the Otto-von-Guericke University Magdeburg, Gommern, Germany
| | - Gerd R Burmester
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Medizinische Klinik für Rheumatologie und Klinische Immunologie, Charitéplatz 1, 10117, Berlin, Germany.
| |
Collapse
|
34
|
Zhang M, Zeng Q, Zhou S, Zhu G, Xu Y, Gao R, Su W, Wang R. Mendelian randomization study on causal association of IL-6 signaling with pulmonary arterial hypertension. Clin Exp Hypertens 2023; 45:2183963. [PMID: 36871578 DOI: 10.1080/10641963.2023.2183963] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Abstract
BACKGROUND A recent Mendelian randomization (MR) did not support an effect of the lead interleukin-6 receptor (IL-6 R) variant on risk of pulmonary arterial hypertension (PAH). Thus, we used two sets of genetic instrumental variants (IVs) and publicly available PAH genome-wide association studies (GWAS) to reassess the genetic causal link between IL-6 signaling and PAH. METHODS Six independent IL-6 signaling and 34 independent soluble IL-6 receptor (sIL-6 R) genetic IVs from recent MR reports and PAH GWAS including 162,962 European individuals were used to perform this two-sample MR study. RESULTS We found that as IL-6 signaling genetically increased, the risk of PAH reduced using IVW (odds ratio [OR] = 0.023, 95% confidence interval [CI]: 0.0013-0.393; p = .0093) and weighted median (OR = 0.033, 95% CI: 0.0024-0.467; p = .0116). Otherwise, as sIL-6 R genetically increased, the risk of PAH increased using IVW (OR = 1.34, 95% CI: 1.16-1.56; p = .0001), weighted median (OR = 1.36, 95% CI: 1.10-1.68; p = .005), MR-Egger (OR = 1.43, 95% CI: 1.05-1.94; p = .03), and weighted mode (OR = 1.35, 95% CI for OR: 1.12-1.63; p = .0035). CONCLUSION Our analysis suggested the causal link between genetically increased sIL-6 R and increased risk of PAH and between genetically increased IL-6 signaling and reduced risk of PAH. Thus, higher sIL-6 R levels may be a risk factor for patients with PAH, whereas higher IL-6 signaling may be a protective factor for patients with PAH.
Collapse
Affiliation(s)
- Min Zhang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, China
| | - Qi Zeng
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, China
| | - Shan Zhou
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, China
| | - Gaizhi Zhu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, China
| | - Yaqi Xu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, China
| | - Ran Gao
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, China
| | - Wenting Su
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, China
| | - Renxi Wang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, China
| |
Collapse
|
35
|
Cook SA. Understanding interleukin 11 as a disease gene and therapeutic target. Biochem J 2023; 480:1987-2008. [PMID: 38054591 PMCID: PMC10754292 DOI: 10.1042/bcj20220160] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 11/13/2023] [Accepted: 11/27/2023] [Indexed: 12/07/2023]
Abstract
Interleukin 11 (IL11) is an elusive member of the IL6 family of cytokines. While initially thought to be a haematopoietic and cytoprotective factor, more recent data show instead that IL11 is redundant for haematopoiesis and toxic. In this review, the reasons that led to the original misunderstandings of IL11 biology, which are now understandable, are explained with particular attention on the use of recombinant human IL11 in mice and humans. Following tissue injury, as part of an evolutionary ancient homeostatic response, IL11 is secreted from damaged mammalian cells to signal via JAK/STAT3, ERK/P90RSK, LKB1/mTOR and GSK3β/SNAI1 in autocrine and paracrine. This activates a program of mesenchymal transition of epithelial, stromal, and endothelial cells to cause inflammation, fibrosis, and stalled endogenous tissue repair, leading to organ failure. The role of IL11 signalling in cell- and organ-specific pathobiology is described, the large unknowns about IL11 biology are discussed and the promise of targeting IL11 signalling as a therapeutic approach is reviewed.
Collapse
Affiliation(s)
- Stuart A. Cook
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore, Singapore
| |
Collapse
|
36
|
El-Husseini ZW, Khalenkow D, Lan A, van der Molen T, Brightling C, Papi A, Rabe KF, Siddiqui S, Singh D, Kraft M, Beghe B, van den Berge M, van Gosliga D, Nawijn MC, Rose-John S, Koppelman GH, Gosens R. An epithelial gene signature of trans-IL-6 signaling defines a subgroup of type 2-low asthma. Respir Res 2023; 24:308. [PMID: 38062491 PMCID: PMC10704725 DOI: 10.1186/s12931-023-02617-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Asthma is stratified into type 2-high and type 2-low inflammatory phenotypes. Limited success has been achieved in developing drugs that target type 2-low inflammation. Previous studies have linked IL-6 signaling to severe asthma. IL-6 cooperates with soluble-IL-6Rα to activate cell signaling in airway epithelium. OBJECTIVE We sought to study the role of sIL-6Rα amplified IL-6 signaling in airway epithelium and to develop an IL-6+ sIL-6Rα gene signature that may be used to select asthma patients who potentially respond to anti-IL-6 therapy. METHODS Human airway epithelial cells were stimulated with combinations of IL-6, sIL-6Rα, and inhibitors, sgp130 (Olamkicept), and anti-IL-6R (Tocilizumab), to assess effects on pathway activation, epithelial barrier integrity, and gene expression. A gene signature was generated to identify IL-6 high patients using bronchial biopsies and nasal brushes. RESULTS Soluble-IL-6Rα amplified the activation of the IL-6 pathway, shown by the increase of STAT3 phosphorylation and stronger gene induction in airway epithelial cells compared to IL-6 alone. Olamkicept and Tocilizumab inhibited the effect of IL-6 + sIL-6Rα on gene expression. We developed an IL-6 + sIL-6Rα gene signature and observed enrichment of this signature in bronchial biopsies but not nasal brushes from asthma patients compared to healthy controls. An IL-6 + sIL-6Rα gene signature score was associated with lower levels of sputum eosinophils in asthma. CONCLUSION sIL-6Rα amplifies IL-6 signaling in bronchial epithelial cells. Higher local airway IL-6 + sIL-6Rα signaling is observed in asthma patients with low sputum eosinophils.
Collapse
Affiliation(s)
- Zaid W El-Husseini
- Department of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
- Department of Molecular Pharmacology, Faculty of Science and Engineering, Groningen Research Institute of Pharmacy, University of Groningen, 9713 AV, Groningen, The Netherlands
| | - Dmitry Khalenkow
- Department of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
| | - Andy Lan
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
- Department of Molecular Pharmacology, Faculty of Science and Engineering, Groningen Research Institute of Pharmacy, University of Groningen, 9713 AV, Groningen, The Netherlands
| | - Thys van der Molen
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
| | - Chris Brightling
- Department of Infection, Immunity and Inflammation, Institute for Lung Health, University of Leicester, Leicester, UK
| | - Alberto Papi
- Department of Respiratory Medicine, University of Ferrara, Ferrara, Italy
| | - Klaus F Rabe
- Department of Medicine, Christian Albrechts University Kiel, Kiel and Lungen Clinic Grosshansdorf (Members of the German Center for Lung Research (DZL)), Grosshansdorf, Germany
| | - Salman Siddiqui
- National Heart and Lung Institute, Imperial College and Imperial NIHR Biomedical Research Centre, London, UK
| | - Dave Singh
- Medicines Evaluation Unit, Manchester University NHS Foundation Hospital Trust, University of Manchester, Manchester, UK
| | - Monica Kraft
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Bianca Beghe
- University of Modena and Reggio Emilia, AOU of Modena, Modena, Italy
| | - Maarten van den Berge
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
- Department of Pulmonary Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Djoke van Gosliga
- Department of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
- Department of Pathology and Medical Biology, Experimental Pulmonary and Inflammatory Research (EXPIRE), University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Martijn C Nawijn
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
- Department of Pathology and Medical Biology, Experimental Pulmonary and Inflammatory Research (EXPIRE), University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | | | - Gerard H Koppelman
- Department of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
| | - Reinoud Gosens
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands.
- Department of Molecular Pharmacology, Faculty of Science and Engineering, Groningen Research Institute of Pharmacy, University of Groningen, 9713 AV, Groningen, The Netherlands.
| |
Collapse
|
37
|
Glass J, Robinson RL, Greenway G, Jones G, Sharma S. Diabetic Müller-Glial-Cell-Specific Il6ra Knockout Mice Exhibit Accelerated Retinal Functional Decline and Thinning of the Inner Nuclear Layer. Invest Ophthalmol Vis Sci 2023; 64:1. [PMID: 38038619 PMCID: PMC10697173 DOI: 10.1167/iovs.64.15.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 11/08/2023] [Indexed: 12/02/2023] Open
Abstract
Purpose Interleukin-6 (IL-6) is implicated in the pathology of diabetic retinopathy (DR). IL-6 trans-signaling via soluble IL-6 receptor (IL-6R) is primarily responsible for its pro-inflammatory functions, whereas cis-signaling via membrane-bound IL-6R is anti-inflammatory. Using a Müller-glial-cell-specific Il6ra-/- mouse, we examined how loss of IL-6 cis-signaling in Müller glial cells (MGCs) affected retinal thinning and electroretinography (ERG) response over 9 months of diabetes. Methods Diabetes was induced in wildtype and knockout mice with streptozotocin (40 mg/kg, daily for 5 days). Spectral domain optical coherence tomography (SD-OCT), ERG, and fundoscopy/fluorescein angiography (FA) were assessed at 2, 6, and 9 months of diabetes. MGCs and bipolar neurons were examined in retinal tissue sections by immunofluorescence. Results Diabetic MGC Il6ra-/- mice had significantly thinner retinas than diabetic wildtype mice at 2 (-7.6 µm), 6 (-12.0 µm), and 9 months (-5.0 µm) of diabetes, as well as significant thinning of the inner nuclear layer (INL). Diabetic MGC Il6ra-/- mice also showed a reduction in scotopic B-wave amplitude and B-wave/A-wave ratio earlier than wildtype diabetic mice. In retinal sections, we found a decrease in bipolar neuronal marker PKCα only in diabetic MGC Il6ra-/- mice, which was significantly lower than both controls and diabetic wildtype mice. Glutamine synthetase, a Müller cell marker, was reduced in both wildtype and MGC Il6ra-/- diabetic mice compared to their respective controls. Conclusions IL-6 cis-signaling in MGCs contributes to maintenance of the INL in diabetes, and loss of the IL-6 receptor reduces MGC-mediated neuroprotection of bipolar neurons in the diabetic retina.
Collapse
Affiliation(s)
- Joshua Glass
- Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, Georgia, United States
| | - Rebekah L. Robinson
- Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, Georgia, United States
| | - Grace Greenway
- Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, Georgia, United States
| | - Garrett Jones
- Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, Georgia, United States
| | - Shruti Sharma
- Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, Georgia, United States
- Culver Vision Discovery Institute, Augusta University, Augusta, Georgia, United States
- Department of Ophthalmology, Augusta University, Augusta, Georgia, United States
| |
Collapse
|
38
|
Hoffman JM, Robinson R, Greenway G, Glass J, Budkin S, Sharma S. Blockade of interleukin-6 trans-signaling prevents mitochondrial dysfunction and cellular senescence in retinal endothelial cells. Exp Eye Res 2023; 237:109721. [PMID: 37956941 PMCID: PMC10759313 DOI: 10.1016/j.exer.2023.109721] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/03/2023] [Accepted: 11/07/2023] [Indexed: 11/20/2023]
Abstract
Interleukin-6 (IL-6) is a multifaceted cytokine implicated in the pathogenesis of diabetic retinopathy (DR). Its activity extends through cis- and trans-signaling (TS) pathways, with cis-signaling limited to specific cell types possessing the membrane-bound IL-6 receptor, while trans-signaling broadly activates various cells without the membrane bound IL-6 receptor, including retinal endothelial cells. In this study, we determined the effects of interleukin-6 trans-signaling on mitochondrial dysfunction and cellular senescence in human retinal endothelial cells (HRECs). HRECs were cultured and treated with IL-6 + soluble IL-6R or Hyper IL-6 to activate trans-signaling, along with sgp130Fc for inhibition. RT-PCR was used to analyze gene expression changes associated with inflammation and senescence. Cellular senescence was assessed using SA β-gal staining. Mitochondrial function was evaluated using Seahorse XFe24 Bioanalyzer. IL-6 trans-signaling induced inflammatory gene expression as indicated by the upregulation of ICAM1, MCP1, and SERPINA3 levels. Additionally, it reduced mitochondrial respiration and oxidative phosphorylation, and these effects were counteracted by sgp130Fc. Moreover, IL-6 trans-signaling led to altered expression of apoptosis-associated genes, including downregulation of FIS1, BCL2, and MCL1, while promoting cellular senescence, a phenomenon mitigated by sgp130Fc. These results not only deepen our understanding of IL-6 in DR but also carry broader implications for age-related diseases and the aging process itself. This study underscores the potential therapeutic value of targeting IL-6 trans-signaling with sgp130Fc as a promising anti-inflammatory approach for DR and potentially other inflammatory conditions. Further in-vivo investigations are warranted to elucidate the function of IL-6 trans-signaling in aging-related pathologies and overall organismal health.
Collapse
Affiliation(s)
- Jessica M Hoffman
- Department of Biological Sciences, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA.
| | - Rebekah Robinson
- Center for Biotechnology and Genomic Medicine, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Grace Greenway
- Center for Biotechnology and Genomic Medicine, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Joshua Glass
- Center for Biotechnology and Genomic Medicine, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Stepan Budkin
- Center for Biotechnology and Genomic Medicine, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Shruti Sharma
- Center for Biotechnology and Genomic Medicine, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA; Department of Ophthalmology, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA.
| |
Collapse
|
39
|
Gesiorowski A, Ettich J, Werner J, Wittich C, Pieper S, Padrini G, Behnke K, Floss DM, Lang PA, Moll JM, Scheller J. Bispecific soluble cytokine receptor-nanobody fusions inhibit Interleukin (IL-)6 trans-signaling and IL-12/23 or tumor necrosis factor (TNF) signaling. J Biol Chem 2023; 299:105343. [PMID: 37838173 PMCID: PMC10652096 DOI: 10.1016/j.jbc.2023.105343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 09/18/2023] [Accepted: 10/06/2023] [Indexed: 10/16/2023] Open
Abstract
At least 0.5% of people in the Western world develop inflammatory bowel disease (IBD). While antibodies that block tumor necrosis factor (TNF) α and Interleukin (IL-)23 have been approved for the treatment of IBD, IL-6 antibodies failed in the phase II clinical trial due to non-tolerable side effects. However, two clinical phase II studies suggest that inhibiting IL-6/soluble IL-6R (sIL-6R)-induced trans-signaling via the cytokine receptor gp130 benefit IBD patients with fewer adverse events. Here we develop inhibitors targeting a combination of IL-6/sIL-6R and TNF or IL-12/IL-23 signaling, named cs130-TNFVHHFc and cs130-IL-12/23VHHFc. Surface plasmon resonance experiments showed that recombinant cs130-TNFVHHFc and cs130-IL-12/23VHHFc bind with high affinity to IL-6/sIL-6R complexes and human TNFα (hTNFα) or IL-12/IL-23, respectively. Immunoprecipitation experiments have verified the higher ordered complex formation of the inhibitors with IL-6/sIL-6R and IL-12. We demonstrated that cs130-TNFVHHFc and cs130-IL-12/23VHHFc block IL-6/sIL-6R trans-signaling-induced proliferation and STAT3 phosphorylation of Ba/F3-gp130 cells, as well as hTNFα- or IL-23-induced signaling, respectively. In conclusion, cs130-TNFVHHFc and cs130-IL-12/23VHHFc represent a class of dimeric and bispecific chimeric cytokine inhibitors that consist of a soluble cytokine receptor fused to anti-cytokine nanobodies.
Collapse
Affiliation(s)
- Annika Gesiorowski
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Julia Ettich
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Julia Werner
- Institute of Molecular Medicine II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Christoph Wittich
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Stephan Pieper
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Giacomo Padrini
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Kristina Behnke
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Doreen M Floss
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Philipp A Lang
- Institute of Molecular Medicine II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Jens M Moll
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany; PROvendis GmbH, Muelheim an der Ruhr, Germany
| | - Jürgen Scheller
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany.
| |
Collapse
|
40
|
Ye W, Wang Q, Zhao L, Wang C, Zhang D, Zhou M, Chen F, Wang W, Zhu Z, Guo W, Liu Y, Zou H, Xue Y. Blockade of IL-11 Trans-Signaling or JAK2/STAT3 Signaling Ameliorates the Profibrotic Effect of IL-11. Immunol Invest 2023; 52:703-716. [PMID: 37401665 DOI: 10.1080/08820139.2023.2222746] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2023]
Abstract
OBJECTIVES Systemic sclerosis (SSc) is a rare rheumatic disease characterized by vascular damage, dysregulated immune response, and fibrosis. Interleukin-11 (IL-11) is upregulated in SSc. This study aimed to investigate the pathological and therapeutic role of the IL-11 trans-signaling pathway in SSc. METHODS Plasma IL-11 level was evaluated in 32 patients with SSc and 15 healthy controls, while the expression levels of ADAM10, ADAM17, IL-11, IL-11 Rα, or IL-11 co-stained with CD3 or CD163 in the skin of SSc patients and healthy controls were analyzed. Fibroblasts were treated with IL-11 and ionomycin to evaluate the profibrotic effect of IL-11 trans-signaling pathway. TJ301 (sgp130Fc) and WP1066 (a JAK2/STAT3 inhibitor) intervention groups were set up to investigate the antifibrotic effect of targeting IL-11. RESULTS Levels of plasma IL-11 were extremely low in most SSc patients and healthy controls. In contrast, levels of IL-11, IL-11 Rα, and ADAM10, but not ADAM17, were significantly elevated in the skin of SSc patients. Moreover, the numbers of IL-11+ CD3+ cells and IL-11+ CD163+ cells were increased in the skin of SSc patients. Besides, IL-11 and ADAM10 were also elevated in the skin and pulmonary of bleomycin-induced SSc mouse. Fibroblasts co-stimulated with IL-11 and ionomycin showed increased expression of COL3 and phosphorylation of STAT3, which could be inhibited by TJ301 or WP1066. TJ301 also ameliorated skin and lung fibrosis in BLM-induced SSc mouse. CONCLUSIONS IL-11 induces fibrosis in SSc by regulating the trans-signaling pathway. Blockage of sgp130Fc or inhibition of the JAK2/STAT3 pathway could ameliorate the profibrotic effect of IL-11.
Collapse
Affiliation(s)
- Wenjing Ye
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Qian Wang
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Li Zhao
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Changcheng Wang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Dandan Zhang
- MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Mengyu Zhou
- MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Fangfang Chen
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Weiguo Wang
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Zaihua Zhu
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Wenyu Guo
- Clinical Development, I-Mab Biopharma, Hangzhou, China
| | - Yun Liu
- MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Hejian Zou
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yu Xue
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
41
|
Orange ST, Leslie J, Ross M, Mann DA, Wackerhage H. The exercise IL-6 enigma in cancer. Trends Endocrinol Metab 2023; 34:749-763. [PMID: 37633799 DOI: 10.1016/j.tem.2023.08.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/28/2023]
Abstract
Interleukin (IL)-6 elicits both anticancer and procancer effects depending on the context, which we have termed the 'exercise IL-6 enigma'. IL-6 is released from skeletal muscles during exercise to regulate short-term energy availability. Exercise-induced IL-6 provokes biological effects that may protect against cancer by improving insulin sensitivity, stimulating the production of anti-inflammatory cytokines, mobilising immune cells, and reducing DNA damage in early malignant cells. By contrast, IL-6 continuously produced by leukocytes in inflammatory sites drives tumorigenesis by promoting chronic inflammation and activating tumour-promoting signalling pathways. How can a molecule have such opposing effects on cancer? Here, we review the roles of IL-6 in chronic inflammation, tumorigenesis, and exercise-associated cancer prevention and define the factors that underpin the exercise IL-6 enigma.
Collapse
Affiliation(s)
- Samuel T Orange
- Newcastle University Centre for Cancer, Newcastle University, Newcastle upon Tyne, UK; School of Biomedical, Nutritional and Sport Sciences, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.
| | - Jack Leslie
- Newcastle University Centre for Cancer, Newcastle University, Newcastle upon Tyne, UK; Newcastle Fibrosis Research Group, Bioscience Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Mark Ross
- Institute of Life and Earth Sciences, School of Energy, Geoscience, Infrastructure and Society, Heriot-Watt University, Edinburgh, UK
| | - Derek A Mann
- Newcastle University Centre for Cancer, Newcastle University, Newcastle upon Tyne, UK; Newcastle Fibrosis Research Group, Bioscience Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Henning Wackerhage
- Department of Sport & Health Science, Technical University of Munich, Munich, Germany
| |
Collapse
|
42
|
Salvioli S, Basile MS, Bencivenga L, Carrino S, Conte M, Damanti S, De Lorenzo R, Fiorenzato E, Gialluisi A, Ingannato A, Antonini A, Baldini N, Capri M, Cenci S, Iacoviello L, Nacmias B, Olivieri F, Rengo G, Querini PR, Lattanzio F. Biomarkers of aging in frailty and age-associated disorders: State of the art and future perspective. Ageing Res Rev 2023; 91:102044. [PMID: 37647997 DOI: 10.1016/j.arr.2023.102044] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/24/2023] [Accepted: 08/25/2023] [Indexed: 09/01/2023]
Abstract
According to the Geroscience concept that organismal aging and age-associated diseases share the same basic molecular mechanisms, the identification of biomarkers of age that can efficiently classify people as biologically older (or younger) than their chronological (i.e. calendar) age is becoming of paramount importance. These people will be in fact at higher (or lower) risk for many different age-associated diseases, including cardiovascular diseases, neurodegeneration, cancer, etc. In turn, patients suffering from these diseases are biologically older than healthy age-matched individuals. Many biomarkers that correlate with age have been described so far. The aim of the present review is to discuss the usefulness of some of these biomarkers (especially soluble, circulating ones) in order to identify frail patients, possibly before the appearance of clinical symptoms, as well as patients at risk for age-associated diseases. An overview of selected biomarkers will be discussed in this regard, in particular we will focus on biomarkers related to metabolic stress response, inflammation, and cell death (in particular in neurodegeneration), all phenomena connected to inflammaging (chronic, low-grade, age-associated inflammation). In the second part of the review, next-generation markers such as extracellular vesicles and their cargos, epigenetic markers and gut microbiota composition, will be discussed. Since recent progresses in omics techniques have allowed an exponential increase in the production of laboratory data also in the field of biomarkers of age, making it difficult to extract biological meaning from the huge mass of available data, Artificial Intelligence (AI) approaches will be discussed as an increasingly important strategy for extracting knowledge from raw data and providing practitioners with actionable information to treat patients.
Collapse
Affiliation(s)
- Stefano Salvioli
- Department of Medical and Surgical Science, University of Bologna, Bologna, Italy; IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.
| | | | - Leonardo Bencivenga
- Department of Translational Medical Sciences, University of Naples Federico II, Napoli, Italy
| | - Sara Carrino
- Department of Medical and Surgical Science, University of Bologna, Bologna, Italy
| | - Maria Conte
- Department of Medical and Surgical Science, University of Bologna, Bologna, Italy
| | - Sarah Damanti
- IRCCS Ospedale San Raffaele and Vita-Salute San Raffaele University, Milano, Italy
| | - Rebecca De Lorenzo
- IRCCS Ospedale San Raffaele and Vita-Salute San Raffaele University, Milano, Italy
| | - Eleonora Fiorenzato
- Parkinson's Disease and Movement Disorders Unit, Center for Rare Neurological Diseases (ERN-RND), Department of Neurosciences, University of Padova, Padova, Italy
| | - Alessandro Gialluisi
- Department of Epidemiology and Prevention, IRCCS NEUROMED, Pozzilli, Italy; EPIMED Research Center, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Assunta Ingannato
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy; IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Angelo Antonini
- Parkinson's Disease and Movement Disorders Unit, Center for Rare Neurological Diseases (ERN-RND), Department of Neurosciences, University of Padova, Padova, Italy; Center for Neurodegenerative Disease Research (CESNE), Department of Neurosciences, University of Padova, Padova, Italy
| | - Nicola Baldini
- IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy; Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Miriam Capri
- Department of Medical and Surgical Science, University of Bologna, Bologna, Italy
| | - Simone Cenci
- IRCCS Ospedale San Raffaele and Vita-Salute San Raffaele University, Milano, Italy
| | - Licia Iacoviello
- Department of Epidemiology and Prevention, IRCCS NEUROMED, Pozzilli, Italy; EPIMED Research Center, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Benedetta Nacmias
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy; IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Fabiola Olivieri
- Department of Clinical and Molecular Sciences, Università Politecnica Delle Marche, Ancona, Italy; Clinic of Laboratory and Precision Medicine, IRCCS INRCA, Ancona, Italy
| | - Giuseppe Rengo
- Department of Translational Medical Sciences, University of Naples Federico II, Napoli, Italy; Istituti Clinici Scientifici Maugeri IRCCS, Scientific Institute of Telese Terme, Telese Terme, Italy
| | | | | |
Collapse
|
43
|
Sui C, Lee W. Role of interleukin 6 and its soluble receptor on the diffusion barrier dysfunction of alveolar tissue. Biomed Microdevices 2023; 25:40. [PMID: 37851124 DOI: 10.1007/s10544-023-00680-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/03/2023] [Indexed: 10/19/2023]
Abstract
During respiratory infection, barrier dysfunction in alveolar tissue can result from "cytokine storm" caused by overly reactive immune response. Particularly, interleukin 6 (IL-6) is implicated as a key biomarker of cytokine storm responsible for and further progression to pulmonary edema. In this study, alveolar-like tissue was reconstructed in a microfluidic device with: (1) human microvascular lung endothelial cells (HULEC-5a) cultured under flow-induced shear stress and (2) human epithelial cells (Calu-3) cultured at air-liquid interface. The effects of IL-6 and the soluble form of its receptor (sIL-6R) on the permeability, electrical resistance, and morphology of the endothelial and epithelial layers were evaluated. The diffusion barrier properties of both the endothelial and epithelial layers were significantly degraded only when IL-6 treatment was combined with sIL-6R. As suggested by recent review and clinical studies, our results provide unequivocal evidence that the barrier dysfunction occurs through trans-signaling in which IL-6 and sIL-6R form a complex and then bind to the surface of endothelial and epithelial cells, but not by classical signaling in which IL-6 binds to membrane-expressed IL-6 receptor. This finding suggests that the role of both IL-6 and sIL-6R should be considered as important biomarkers in developing strategies for treating cytokine storm.
Collapse
Affiliation(s)
- Chao Sui
- Department of Chemical Engineering and Materials Science, Stevens Institute of Technology, 1 Castle Point On Hudson, Hoboken, New Jersey, 07030, USA
| | - Woo Lee
- Department of Chemical Engineering and Materials Science, Stevens Institute of Technology, 1 Castle Point On Hudson, Hoboken, New Jersey, 07030, USA.
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, 1 Castle Point On Hudson, Hoboken, New Jersey, 07030, USA.
| |
Collapse
|
44
|
Sun T, Chen Q, Mei J, Li Y. Associations between serum estradiol and IL-6/sIL-6R/sgp130 complex in female patients with major depressive disorder. BMC Psychiatry 2023; 23:742. [PMID: 37828513 PMCID: PMC10568828 DOI: 10.1186/s12888-023-05248-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 10/03/2023] [Indexed: 10/14/2023] Open
Abstract
BACKGROUND It has been hypothesized that the IL-6/sIL-6R/sgp130 complex, an inflammatory complex, plays a critical role in the pathogenesis of major depressive disorder (MDD). Estradiol (E2) is a sex steroid hormone involved in emotional regulation and MDD. This study aimed to investigate the relationship between E2 and IL-6/sIL-6R/sgp130 complex in patients with MDD. METHODS Using enzyme-linked immunosorbent assay, the levels of IL-6, sIL-6Rα, and sgp130 were compared between 117 female patients with MDD and 122 healthy controls.The serum concentrations of E2 and other biomarkers were also measured. RESULTS (1) The serum levels of IL-6 and sIL-6Rα in patients with MDD were significantly higher than those in the control group, while the serum levels of sgp130 and E2 were significantly lower (all P < 0.05). (2) Low levels of E2 were associated with high levels of IL-6 and low levels of sgp130 (all P < 0.01). (3) HAMD-24 score was positively correlated with the serum level of IL-6, but negatively correlated with the serum levels of sgp130 and E2(all P < 0.05). (4) IL-6 and sgp130 had certain prognostic values in MDD, and the combination of various indicators showed a significantly superior prognostic value. CONCLUSIONS The IL6/sIL-6R/sgp130 complex in female patients with MDD was closely related to E2 level. In addition, IL-6 and sgp130 may be valuable serum biomarkers for the diagnosis and prognosis of MDD in women.
Collapse
Affiliation(s)
- Ting Sun
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Zhangzhidong Road, Wuchang District, Wuhan, 430060, Hubei, China
| | - Qian Chen
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Zhangzhidong Road, Wuchang District, Wuhan, 430060, Hubei, China
| | - Junchi Mei
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Zhangzhidong Road, Wuchang District, Wuhan, 430060, Hubei, China
| | - Yan Li
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Zhangzhidong Road, Wuchang District, Wuhan, 430060, Hubei, China.
| |
Collapse
|
45
|
Rose-John S, Jenkins BJ, Garbers C, Moll JM, Scheller J. Targeting IL-6 trans-signalling: past, present and future prospects. Nat Rev Immunol 2023; 23:666-681. [PMID: 37069261 PMCID: PMC10108826 DOI: 10.1038/s41577-023-00856-y] [Citation(s) in RCA: 197] [Impact Index Per Article: 98.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/01/2023] [Indexed: 04/19/2023]
Abstract
Interleukin-6 (IL-6) is a key immunomodulatory cytokine that affects the pathogenesis of diverse diseases, including autoimmune diseases, chronic inflammatory conditions and cancer. Classical IL-6 signalling involves the binding of IL-6 to the membrane-bound IL-6 receptor α-subunit (hereafter termed 'mIL-6R') and glycoprotein 130 (gp130) signal-transducing subunit. By contrast, in IL-6 trans-signalling, complexes of IL-6 and the soluble form of IL-6 receptor (sIL-6R) signal via membrane-bound gp130. A third mode of IL-6 signalling - known as cluster signalling - involves preformed complexes of membrane-bound IL-6-mIL-6R on one cell activating gp130 subunits on target cells. Antibodies and small molecules have been developed that block all three forms of IL-6 signalling, but in the past decade, IL-6 trans-signalling has emerged as the predominant pathway by which IL-6 promotes disease pathogenesis. The first selective inhibitor of IL-6 trans-signalling, sgp130, has shown therapeutic potential in various preclinical models of disease and olamkicept, a sgp130Fc variant, had promising results in phase II clinical studies for inflammatory bowel disease. Technological developments have already led to next-generation sgp130 variants with increased affinity and selectivity towards IL-6 trans-signalling, along with indirect strategies to block IL-6 trans-signalling. Here, we summarize our current understanding of the biological outcomes of IL-6-mediated signalling and the potential for targeting this pathway in the clinic.
Collapse
Affiliation(s)
- Stefan Rose-John
- Biochemical Institute, Medical Faculty, Christian-Albrechts-University, Kiel, Germany
| | - Brendan J Jenkins
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Science, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia
| | - Christoph Garbers
- Department of Pathology, Otto-von-Guericke-University Magdeburg, Medical Faculty, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation (GC:I3), Otto-von-Guericke-University, Magdeburg, Germany
- Center for Health and Medical Prevention (CHaMP), Otto-von-Guericke-University, Magdeburg, Germany
| | - Jens M Moll
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Jürgen Scheller
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany.
| |
Collapse
|
46
|
Ren J, Wang XQ, Nakao T, Libby P, Shi GP. Differential Roles of Interleukin-6 in Severe Acute Respiratory Syndrome-Coronavirus-2 Infection and Cardiometabolic Diseases. CARDIOLOGY DISCOVERY 2023; 3:166-182. [PMID: 38152628 PMCID: PMC10750760 DOI: 10.1097/cd9.0000000000000096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
Severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) infection can lead to a cytokine storm, unleashed in part by pyroptosis of virus-infected macrophages and monocytes. Interleukin-6 (IL-6) has emerged as a key participant in this ominous complication of COVID-19. IL-6 antagonists have improved outcomes in patients with COVID-19 in some, but not all, studies. IL-6 signaling involves at least 3 distinct pathways, including classic-signaling, trans-signaling, and trans-presentation depending on the localization of IL-6 receptor and its binding partner glycoprotein gp130. IL-6 has become a therapeutic target in COVID-19, cardiovascular diseases, and other inflammatory conditions. However, the efficacy of inhibition of IL-6 signaling in metabolic diseases, such as obesity and diabetes, may depend in part on cell type-dependent actions of IL-6 in controlling lipid metabolism, glucose uptake, and insulin sensitivity owing to complexities that remain to be elucidated. The present review sought to summarize and discuss the current understanding of how and whether targeting IL-6 signaling ameliorates outcomes following SARS-CoV-2 infection and associated clinical complications, focusing predominantly on metabolic and cardiovascular diseases.
Collapse
Affiliation(s)
- Jingjing Ren
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115
| | - Xiao-Qi Wang
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115
| | - Tetsushi Nakao
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115
| | - Peter Libby
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115
| | - Guo-Ping Shi
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115
| |
Collapse
|
47
|
Lin W, Song H, Shen J, Wang J, Yang Y, Yang Y, Cao J, Xue L, Zhao F, Xiao T, Lin R. Functional role of skeletal muscle-derived interleukin-6 and its effects on lipid metabolism. Front Physiol 2023; 14:1110926. [PMID: 37555019 PMCID: PMC10405179 DOI: 10.3389/fphys.2023.1110926] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 07/06/2023] [Indexed: 08/10/2023] Open
Abstract
The detrimental impact of obesity on human health is increasingly evident with the rise in obesity-related diseases. Skeletal muscle, the crucial organ responsible for energy balance metabolism, plays a significant role as a secretory organ by releasing various myokines. Among these myokines, interleukin 6 (IL-6) is closely associated with skeletal muscle contraction. IL-6 triggers the process of lipolysis by mobilizing energy-storing adipose tissue, thereby providing energy for physical exercise. This phenomenon also elucidates the health benefits of regular exercise. However, skeletal muscle and adipose tissue maintain a constant interaction, both directly and indirectly. Direct interaction occurs through the accumulation of excess fat within skeletal muscle, known as ectopic fat deposition. Indirect interaction takes place when adipose tissue is mobilized to supply the energy for skeletal muscle during exercise. Consequently, maintaining a functional balance between skeletal muscle and adipose tissue becomes paramount in regulating energy metabolism and promoting overall health. IL-6, as a representative cytokine, participates in various inflammatory responses, including non-classical inflammatory responses such as adipogenesis. Skeletal muscle influences adipogenesis through paracrine mechanisms, primarily by secreting IL-6. In this research paper, we aim to review the role of skeletal muscle-derived IL-6 in lipid metabolism and other physiological activities, such as insulin resistance and glucose tolerance. By doing so, we provide valuable insights into the regulatory function of skeletal muscle-derived myokines in lipid metabolism.
Collapse
Affiliation(s)
- Weimin Lin
- *Correspondence: Weimin Lin, ; Ruiyi Lin,
| | | | | | | | | | | | | | | | | | | | - Ruiyi Lin
- College of Animal Sciences (College of Bee Science), Fujian Agriculture and Forestry University, Fuzhou, China
| |
Collapse
|
48
|
de Baat A, Trinh B, Ellingsgaard H, Donath MY. Physiological role of cytokines in the regulation of mammalian metabolism. Trends Immunol 2023:S1471-4906(23)00110-2. [PMID: 37423882 DOI: 10.1016/j.it.2023.06.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/09/2023] [Accepted: 06/09/2023] [Indexed: 07/11/2023]
Abstract
The innate cytokine system is involved in the response to excessive food intake. In this review, we highlight recent advances in our understanding of the physiological role of three prominent cytokines, interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF), in mammalian metabolic regulation. This recent research highlights the pleiotropic and context-dependent functions in the immune-metabolic interplay. IL-1β is activated in response to overloaded mitochondrial metabolism, stimulates insulin secretion, and allocates energy to immune cells. IL-6 is released by contracting skeletal muscle and adipose tissue and directs energy from storing tissues to consuming tissues. TNF induces insulin resistance and prevents ketogenesis. Additionally, the therapeutic potential of modulating the activity of each cytokine is discussed.
Collapse
Affiliation(s)
- Axel de Baat
- Clinic of Endocrinology, Diabetes and Metabolism University Hospital Basel, Basel, Switzerland; Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Beckey Trinh
- The Centre for Physical Activity Research, Rigshospitalet, Copenhagen, Denmark
| | - Helga Ellingsgaard
- The Centre for Physical Activity Research, Rigshospitalet, Copenhagen, Denmark
| | - Marc Y Donath
- Clinic of Endocrinology, Diabetes and Metabolism University Hospital Basel, Basel, Switzerland; Department of Biomedicine, University of Basel, Basel, Switzerland.
| |
Collapse
|
49
|
Xiao R, Lei C, Zhang Y, Zhang M. Interleukin-6 in retinal diseases: From pathogenesis to therapy. Exp Eye Res 2023:109556. [PMID: 37385535 DOI: 10.1016/j.exer.2023.109556] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 06/03/2023] [Accepted: 06/25/2023] [Indexed: 07/01/2023]
Abstract
Interleukin-6 (IL-6) is a pleiotropic cytokine that participates in immunomodulation, inflammation, increases vascular permeability, hematopoiesis, and stimulates cell proliferation, among other biological processes. It exerts effects primarily through the classic and trans-signaling pathways. Many studies have demonstrated that IL-6 plays a critical role in the development of retinal diseases including diabetic retinopathy, uveitis, age-related macular degeneration, glaucoma, retinal vein occlusion, central serous chorioretinopathy and proliferative vitreoretinopathy. Thus, the progressive development of drugs targeting IL-6 and IL-6 receptor may play a role in the treatment of multiple retinal diseases. In this article, we comprehensively review the IL-6's biological functions of and its mechanisms in the pathogenesis of various retinal diseases. Furthermore, we summarize the drugs targeting IL-6 and its receptor and prospect their potential application in retinal diseases, hoping to provide new ideas for the treatment of retinal diseases.
Collapse
Affiliation(s)
- Ruihan Xiao
- Department of Ophthalmology and Research Laboratory of Macular Disease, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chunyan Lei
- Department of Ophthalmology and Research Laboratory of Macular Disease, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yi Zhang
- Department of Ophthalmology and Research Laboratory of Macular Disease, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Meixia Zhang
- Department of Ophthalmology and Research Laboratory of Macular Disease, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
50
|
Gatsiou A, Tual-Chalot S, Napoli M, Ortega-Gomez A, Regen T, Badolia R, Cesarini V, Garcia-Gonzalez C, Chevre R, Ciliberti G, Silvestre-Roig C, Martini M, Hoffmann J, Hamouche R, Visker JR, Diakos N, Wietelmann A, Silvestris DA, Georgiopoulos G, Moshfegh A, Schneider A, Chen W, Guenther S, Backs J, Kwak S, Selzman CH, Stamatelopoulos K, Rose-John S, Trautwein C, Spyridopoulos I, Braun T, Waisman A, Gallo A, Drakos SG, Dimmeler S, Sperandio M, Soehnlein O, Stellos K. The RNA editor ADAR2 promotes immune cell trafficking by enhancing endothelial responses to interleukin-6 during sterile inflammation. Immunity 2023; 56:979-997.e11. [PMID: 37100060 DOI: 10.1016/j.immuni.2023.03.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 01/02/2023] [Accepted: 03/30/2023] [Indexed: 04/28/2023]
Abstract
Immune cell trafficking constitutes a fundamental component of immunological response to tissue injury, but the contribution of intrinsic RNA nucleotide modifications to this response remains elusive. We report that RNA editor ADAR2 exerts a tissue- and stress-specific regulation of endothelial responses to interleukin-6 (IL-6), which tightly controls leukocyte trafficking in IL-6-inflamed and ischemic tissues. Genetic ablation of ADAR2 from vascular endothelial cells diminished myeloid cell rolling and adhesion on vascular walls and reduced immune cell infiltration within ischemic tissues. ADAR2 was required in the endothelium for the expression of the IL-6 receptor subunit, IL-6 signal transducer (IL6ST; gp130), and subsequently, for IL-6 trans-signaling responses. ADAR2-induced adenosine-to-inosine RNA editing suppressed the Drosha-dependent primary microRNA processing, thereby overwriting the default endothelial transcriptional program to safeguard gp130 expression. This work demonstrates a role for ADAR2 epitranscriptional activity as a checkpoint in IL-6 trans-signaling and immune cell trafficking to sites of tissue injury.
Collapse
Affiliation(s)
- Aikaterini Gatsiou
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK; RNA Metabolism and Vascular Inflammation Laboratory, Institute of Cardiovascular Regeneration and Department of Cardiology, JW Goethe University Frankfurt, Frankfurt am Main, Germany.
| | - Simon Tual-Chalot
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Matteo Napoli
- Institute for Cardiovascular Physiology and Pathophysiology, Walter Brendel Center for Experimental Medicine Biomedical Center (BMC), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Almudena Ortega-Gomez
- Institute for Cardiovascular Prevention (IPEK), LMU Munich Hospital, Munich, Germany
| | - Tommy Regen
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Rachit Badolia
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Valeriana Cesarini
- Department of Pediatric Hematology/Oncology and Cellular and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | - Raphael Chevre
- Institute for Cardiovascular Prevention (IPEK), LMU Munich Hospital, Munich, Germany; Institute for Experimental Pathology (ExPat), Center for Molecular Biology of Inflammation, WWU Muenster, Muenster, Germany
| | - Giorgia Ciliberti
- Department of Cardiovascular Research, European Center for Angioscience (ECAS), Heidelberg University, Mannheim, Germany
| | - Carlos Silvestre-Roig
- Institute for Cardiovascular Prevention (IPEK), LMU Munich Hospital, Munich, Germany; Institute for Experimental Pathology (ExPat), Center for Molecular Biology of Inflammation, WWU Muenster, Muenster, Germany
| | - Maurizio Martini
- Fondazione Policlinico Universitario "A. Gemelli," IRCCS, UOC Anatomia Patologica, Rome, Italy; Istituto di Anatomia Patologica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Jedrzej Hoffmann
- Department of Cardiology, Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Rana Hamouche
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Joseph R Visker
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Nikolaos Diakos
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Astrid Wietelmann
- Max-Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Domenico Alessandro Silvestris
- Department of Pediatric Hematology/Oncology and Cellular and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Georgios Georgiopoulos
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece; Translational Research Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Ali Moshfegh
- Kancera AB, Stockholm, Sweden; Department of Oncology and Pathology at Karolinska Institutet, Stockholm, Sweden
| | - Andre Schneider
- Max-Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Wei Chen
- Department of Biology, Southern University of Science and Technology, Shenzhen, Guangdong, China; Medi-X Institute, SUSTech Academy for Advanced Interdisciplinary Studies, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Stefan Guenther
- Max-Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Johannes Backs
- Institute of Experimental Cardiology, University Hospital Heidelberg, Heidelberg, Germany; German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung, DZHK), Heidelberg/Mannheim Partner Site, Heidelberg and Mannheim, Germany
| | - Shin Kwak
- Department of Molecular Neuropathogenesis, Tokyo Medical University, Tokyo, Japan
| | - Craig H Selzman
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, Salt Lake City, UT, USA; Division of Cardiothoracic Surgery, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Kimon Stamatelopoulos
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece; Translational Research Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Stefan Rose-John
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Christian Trautwein
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Ioakim Spyridopoulos
- Translational Research Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK; Department of Cardiology, Freeman Hospital, Newcastle upon Tyne NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Thomas Braun
- Max-Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Angela Gallo
- Department of Pediatric Hematology/Oncology and Cellular and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Stavros G Drakos
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, Salt Lake City, UT, USA; Division of Cardiovascular Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Stefanie Dimmeler
- Institute of Cardiovascular Regeneration, Center of Molecular Medicine, JW Goethe University Frankfurt, Frankfurt am Main, Germany; German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung, DZHK), Frankfurt Partner Site, Germany
| | - Markus Sperandio
- Institute for Cardiovascular Physiology and Pathophysiology, Walter Brendel Center for Experimental Medicine Biomedical Center (BMC), Ludwig-Maximilians-Universität München, Munich, Germany; German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung, DZHK), Munich Heart Alliance Partner Site, Munich, Germany
| | - Oliver Soehnlein
- Institute for Cardiovascular Prevention (IPEK), LMU Munich Hospital, Munich, Germany; Institute for Experimental Pathology (ExPat), Center for Molecular Biology of Inflammation, WWU Muenster, Muenster, Germany; German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung, DZHK), Munich Heart Alliance Partner Site, Munich, Germany; Department of Physiology and Pharmacology (FyFa), Karolinska Institutet, Stockholm, Sweden
| | - Konstantinos Stellos
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK; RNA Metabolism and Vascular Inflammation Laboratory, Institute of Cardiovascular Regeneration and Department of Cardiology, JW Goethe University Frankfurt, Frankfurt am Main, Germany; Department of Cardiovascular Research, European Center for Angioscience (ECAS), Heidelberg University, Mannheim, Germany; German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung, DZHK), Heidelberg/Mannheim Partner Site, Heidelberg and Mannheim, Germany; Cardio-Pulmonary Institute (CPI), Frankfurt am Main, Germany.
| |
Collapse
|